1
|
Poddar S, Chauvin SD, Archer CH, Qian W, Castillo-Badillo JA, Yin X, Disbennett WM, Miner CA, Holley JA, Naismith TV, Stinson WA, Wei X, Ning Y, Fu J, Ochoa TA, Surve N, Zaver SA, Wodzanowski KA, Balka KR, Venkatraman R, Liu C, Rome K, Bailis W, Shiba Y, Cherry S, Shin S, Semenkovich CF, De Nardo D, Yoh S, Roberson EDO, Chanda SK, Kast DJ, Miner JJ. ArfGAP2 promotes STING proton channel activity, cytokine transit, and autoinflammation. Cell 2025; 188:1605-1622.e26. [PMID: 39947179 PMCID: PMC11928284 DOI: 10.1016/j.cell.2025.01.027] [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: 04/06/2024] [Revised: 11/03/2024] [Accepted: 01/17/2025] [Indexed: 02/23/2025]
Abstract
Stimulator of interferon genes (STING) transmits signals downstream of the cytosolic DNA sensor cyclic guanosine monophosphate-AMP synthase (cGAS), leading to transcriptional upregulation of cytokines. However, components of the STING signaling pathway, such as IRF3 and IFNAR1, are not essential for autoinflammatory disease in STING gain-of-function (STING-associated vasculopathy with onset in infancy [SAVI]) mice. Recent discoveries revealed that STING also functions as a proton channel that deacidifies the Golgi apparatus. Because pH impacts Golgi enzyme activity, protein maturation, and trafficking, we hypothesized that STING proton channel activity influences multiple Golgi functions. Here, we show that STING-mediated proton efflux non-transcriptionally regulates Golgi trafficking of protein cargos. This process requires the Golgi-associated protein ArfGAP2, a cell-type-specific dual regulator of STING-mediated proton efflux and signaling. Deletion of ArfGAP2 in hematopoietic and endothelial cells markedly reduces STING-mediated cytokine and chemokine secretion, immune cell activation, and autoinflammatory pathology in SAVI mice. Thus, ArfGAP2 facilitates STING-mediated signaling and cytokine release in hematopoietic cells, significantly contributing to autoinflammatory disease pathogenesis.
Collapse
Affiliation(s)
- Subhajit Poddar
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Samuel D Chauvin
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Christopher H Archer
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Wei Qian
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Jean A Castillo-Badillo
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Xin Yin
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA; State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - W Miguel Disbennett
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Cathrine A Miner
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Joe A Holley
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Teresa V Naismith
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - W Alexander Stinson
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Xiaochao Wei
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Yue Ning
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jiayuan Fu
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Trini A Ochoa
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Nehalee Surve
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Shivam A Zaver
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kimberly A Wodzanowski
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Katherine R Balka
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Rajan Venkatraman
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Canyu Liu
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kelly Rome
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Will Bailis
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yoko Shiba
- Faculty of Science and Engineering, Iwate University, Morioka 020-8551, Japan
| | - Sara Cherry
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sunny Shin
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Clay F Semenkovich
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Dominic De Nardo
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Sunnie Yoh
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Elisha D O Roberson
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Sumit K Chanda
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - David J Kast
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA.
| | - Jonathan J Miner
- Division of Rheumatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA; Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Colton Center for Autoimmunity, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
2
|
Cao JF, Yang GJ, Zhang YA, Chen J. Contribution of interleukins in the regulation of teleost fish immunity: A review from the perspective of regulating macrophages. FISH & SHELLFISH IMMUNOLOGY 2025; 158:110173. [PMID: 39909123 DOI: 10.1016/j.fsi.2025.110173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Interleukins (ILs) are potent secreted regulators of a wide range of cell types and cellular activities, particularly in the immune system. They are able to participate in intercellular communication in homeostasis and disease, thereby exerting immune functions. Macrophages serve as the innate immune cells of vertebrates and play a pivotal role in defending against and eliminating external pathogens. In mammals, the immune response mounted by macrophages is intricately linked to ILs. Given the fact that teleost fish have evolved an innate immune system that closely resembles those of mammals, particularly in terms of the functionality of macrophages, raises the intriguing possibility that the regulatory function of ILs in macrophage-mediated immunity might be evolutionarily conserved across both mammal and teleost fish lineages. Consequently, from the perspective of interleukin regulation of macrophages, this review outlines the relationship between ILs and macrophages in teleost fish, and elucidates the regulatory role of ILs of immune cell function in teleost fish, thereby contributing to our understanding of the key role of these cytokines in the prevention and control of aquaculture diseases.
Collapse
Affiliation(s)
- Jia-Feng Cao
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315832, China
| | - Guan-Jun Yang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315832, China
| | - Yong-An Zhang
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Jiong Chen
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315832, China.
| |
Collapse
|
3
|
Yang X, Gan Y, Zhang M, Xie S, Lin M, Zhong L, Song M, Wang J, Huang Y. Transcriptome analysis unveils the mechanisms of oxidative stress, immunotoxicity and neurotoxicity induced by benzotriazole UV stabilizer-328 in zebrafish embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117822. [PMID: 39884018 DOI: 10.1016/j.ecoenv.2025.117822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
As an emerging pollutant, ultraviolet stabilizer-328 (UV-328) has been frequently detected in aquatic environments and attracted great attention. Nevertheless, the toxicity and mechanisms of UV-328 to aquatic organisms are still not fully understood. In particular, the immunotoxicity and neurotoxicity of UV-328 to aquatic organisms and their mechanisms have not been reported yet. In this experiment, the developmental toxicity, oxidative stress, apoptosis, immunotoxicity and neurotoxicity in zebrafish embryos exposed to UV-328 with concentrations of 0.01, 0.1, 1, 10 and 100 µg/L for 120 h were studied. By measuring the growth and developmental indices, production of ROS, enzyme activities, MDA content and expression of genes related to oxidative, immune and nerve, and histopathological analysis, it was found that UV-328 had developmental toxicity to zebrafish larvae, and could induce oxidative stress, immunotoxicity and neurotoxicity to zebrafish larvae even at environmental concentrations with concentration-dependent effects. Moreover, the results of transcriptome analysis and qRT-PCR validation suggested that immune and nerve disorders were caused by UV-328 in zebrafish larvae through regulating the RIG-I-like receptor signaling pathway and neuroactive ligand-receptor interaction, respectively. In addition, transcriptome analysis further revealed that UV-328 could mediate the RIG-I to induce oxidative stress through p38-MAPK/p53 signaling pathway, leading to apoptosis and oxidative damage. In addition, the p38-MAPK signaling pathway enhanced ROS production and activated inflammatory cytokines to induce immunotoxicity. The results of the present work provided important information for understanding the toxicity of UV-328 to aquatic organisms and evaluating its ecological risk in aquatic environment.
Collapse
Affiliation(s)
- Xinlu Yang
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Yijing Gan
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Menghuan Zhang
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Shaolin Xie
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Mingfu Lin
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Lixiang Zhong
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Mengke Song
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, China
| | - Jun Wang
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China.
| | - Yumei Huang
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
4
|
Lin J, Liu S, Xue X, Lv J, Zhao L, Yu L, Wang H, Chen J. Injectable Genetic Engineering Hydrogel for Promoting Spatial Tolerance of Transplanted Kidney in Situ. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2408631. [PMID: 39498870 DOI: 10.1002/advs.202408631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/23/2024] [Indexed: 11/07/2024]
Abstract
The establishment of a tolerant space to realize the co-stimulation of cytokines and contact-dependent molecules remain challenging in allotransplant. Here, an injectable genetically engineered hydrogel (iGE-Gel) is reported, which developed with a multivalent network of FOXP3 engineered extracellular vesicles (Foe-EVs) through the hydrophobic interaction between stearic acid modified hyaluronic acid (HASA) and the membrane phospholipids of extracellular vesicles (EVs). The iGE-Gel exhibited self-healing properties, injectability and biocompatibility. It is revealed that iGE-Gel displayed with abundant regulatory cytokines and coinhibitory contact molecules, promoting the formation of immune tolerance in situ. The multiplex immunohistofluorescence confirmed tolerant niches is dominated by FOXP3+ Tregs and PDL1+ cells in the allograft, which reduced the drainage of alloantigens to subcapsular sinus of lymph nodes, and suppressed the formation of germinal centers. Remarkably, the proportion of alloreactive T cells (IFN-γ/IL-2) and B cells (IgG1/IgG2a/IgG3) as well as the serum titers of donor specific antibody (DSA) is decreased by iGE-Gel. In murine allogeneic transplantation, the injection of iGE-Gel significantly alleviated immune cell infiltration and complement damage in the graft, preserved the structure and function of renal cells and prolonged recipient survival period from 30.8 to 79.3 days, highlighting the potential of iGE-Gel as a transformative treatment in allotransplant.
Collapse
Affiliation(s)
- Jinwen Lin
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| | - Shuaihui Liu
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| | - Xing Xue
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
| | - Junhao Lv
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| | - Lingfei Zhao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| | - Liqin Yu
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| | - Huiping Wang
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| |
Collapse
|
5
|
Sakwe NI, Vuong NB, Black PJ, Ball DD, Thomas P, Beasley HK, Hinton A, Ochieng J, Sakwe AM. Annexin A6 modulates the secretion of pro-inflammatory cytokines and exosomes via interaction with SNAP23 in triple negative breast cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619710. [PMID: 39484394 PMCID: PMC11527025 DOI: 10.1101/2024.10.22.619710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Pro-inflammatory cytokines are secreted via the classical pathway from secretory vesicles or the non-classical pathway via extracellular vesicles (EVs), that together, play critical roles in triple-negative breast cancer (TNBC) progression. Annexin A6 (AnxA6) is a Ca 2+ -dependent membrane-binding protein that in TNBC is implicated in cell growth and invasiveness. AnxA6 is associated with EVs, but whether it affects the secretion of proinflammatory cytokines and/or EVs remains to be fully elucidated. To assess if AnxA6 influences the secretion of cytokines and extracellular vesicles, we used cytokine arrays to analyze secreted factors in cleared culture supernatants from control AnxA6 expressing and AnxA6 downregulated MDA-MB-468 TNBC cells. This revealed the diminished secretion of monocyte chemoattractant protein 1 (MCP-1/CCL2), interleukin 8 (IL-8), dickkopf1 (DKK1), throbospondin-1 (TSP-1), and osteopontin (OPN) following AnxA6 downregulation. We also show that the secretion of small EVs is strongly reduced in AnxA6 downregulated cells and that upregulation of AnxA6 promoted the secretion of treatment was also associated with increased EVs associated Rab7, cholesterol, and MCP-1 levels. Moreover, cholesterol content in EVs was significantly higher in AnxA6-expressing cells than in AnxA6 downregulated cells and following chronic lapatinib induced upregulation of AnxA6. Mechanistically, we demonstrate that the secretion of MCP-1 and/or EVs is AnxA6 dependent and that this requires the translocation of AnxA6 to cellular membranes and its interaction with SNAP23. AnxA6 neutralizing antibodies strongly diminished the survival of AnxA6 low TNBC cells but had minimal effects on the survival of TNBC cells expressing relatively high levels of the protein. Together, these data suggest that AnxA6 facilitates the secretion of EVs and proinflammatory cytokines that may be critical for TNBC progression.
Collapse
|
6
|
Xu Y, Yang T, Xu Q, Tang Y, Yang Q. Vesicle-associated membrane protein 8 knockdown exerts anti-proliferative, pro-apoptotic, anti-autophagic, and pro-ferroptotic effects on colorectal cancer cells by inhibition of the JAK/STAT3 pathway. J Bioenerg Biomembr 2024; 56:419-431. [PMID: 38720136 DOI: 10.1007/s10863-024-10019-w] [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: 03/14/2024] [Accepted: 04/20/2024] [Indexed: 07/03/2024]
Abstract
Vesicle-associated membrane protein 8 (VAMP8), a soluble n-ethylmaleimide-sensitive factor receptor protein, acts as an oncogenic gene in the progression of several malignancies. Nevertheless, the roles and mechanisms of VAMP8 in colorectal cancer (CRC) progression remain unknown. The expression and prognostic significance of VAMP8 in CRC samples were analyzed through bioinformatics analyses. Cell proliferation was detected using CCK-8 and EdU incorporation assays and apoptosis was evaluated via flow cytometry. Western blot analysis was conducted to examine the protein expression. Ferroptosis was evaluated by measurement of iron metabolism, lipid peroxidation, and glutathione (GSH) content. VAMP8 was increased in CRC samples relative to normal samples on the basis of GEPIA and HPA databases. CRC patients with high level of VAMP8 had a worse overall survival. VAMP8 depletion led to a suppression of proliferation and promotion of apoptosis in CRC cells. Additionally, VAMP8 knockdown suppressed beclin1 expression and LC3-II/LC3-I ratio, elevated p62 expression, increased Fe2+, labile iron pool, lipid reactive oxygen species, and malondialdehyde levels, and repressed GSH content and glutathione peroxidase activity. Moreover, VAMP8 knockdown inhibited the activation of janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) pathway in CRC cells. Mechanistically, activation of the JAK/STAT3 pathway by JAK1 or JAK2 overexpression attenuated VAMP8 silencing-mediated anti-proliferative, pro-apoptotic, anti-autophagic, and pro-ferroptotic effects on CRC cells. In conclusion, VAMP8 knockdown affects the proliferation, apoptosis, autophagy, and ferroptosis by the JAK/STAT3 pathway in CRC cells.
Collapse
Affiliation(s)
- Yi Xu
- Department of General Surgery, Nanyang First People's Hospital, Nanyang, China
| | - Tianyao Yang
- Department of General Surgery, Tiantai People's Hospital of Zhejiang Province, Taizhou, China
| | - Qiu Xu
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital, Nanyang, China
- Nanyang Key Laboratory of Thyroid Tumor Prevention and Treatment, Nanyang First People's Hospital, Nanyang, China
| | - Yan Tang
- Department of General Surgery, Nanyang First People's Hospital, Nanyang, China
| | - Qiong Yang
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
7
|
Fiolka R, Wylęgała E, Toborek M, Adamczyk-Zostawa J, Czuba ZP, Wylęgała A. Selective Upregulation of Interleukin 1 Receptor Antagonist and Interleukin-8 in Fuchs' Endothelial Corneal Dystrophy with Accompanying Cataract. J Clin Med 2024; 13:2815. [PMID: 38792359 PMCID: PMC11121793 DOI: 10.3390/jcm13102815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: Patients with Fuchs' endothelial corneal dystrophy (FECD) may have coexisting cataracts and, therefore, may require a cataract surgery, which poses challenges due to potential endothelial cell damage. FECD is a degenerative eye disease of unclear etiology, with inflammatory cytokines maybe playing an important role in its development and progression. The present study aimed to investigate the cytokine profile in the aqueous humor of FECD eyes with cataract. (2) Methods: Fifty-two patients were included in the study, 26 with FECD + cataract and 26 with cataract as a control group. Samples of the aqueous humor were analyzed for pro- and anti-inflammatory cytokines using a Bio-Plex 200 system. (3) Results: Interleukin 1 receptor antagonist (IL-1Ra) and interleukin IL-8 levels were significantly higher in the aqueous humor of FECD + cataract patients compared to the control/cataract group. Moreover, the levels of anti-inflammatory IL-10 showed a strong trend to be higher in the FECD + cataract group compared to the control group. In contrast, there were no statistically significant differences in IL-1β, IL-6, IL-4, IL-10, IL-13, IL-17A, and tumor necrosis factor TNF-α between the groups. (4) Conclusions: Presented research contributes to a better understanding of FECD pathogenesis. Elevated levels of IL-1Ra and IL-8 may serve as a defense mechanism in people with FECD and coexisting cataract.
Collapse
Affiliation(s)
- Rafał Fiolka
- Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland; (E.W.); (J.A.-Z.)
- Doctoral School of the Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Edward Wylęgała
- Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland; (E.W.); (J.A.-Z.)
| | - Michał Toborek
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, FL 33136, USA;
| | - Jowita Adamczyk-Zostawa
- Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland; (E.W.); (J.A.-Z.)
- Doctoral School of the Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Zenon P. Czuba
- Department of Microbiology and Immunology, Faculty of Medical Science, Zabrze Medical University of Silesia in Katowice, 40-055 Katowice, Poland;
| | - Adam Wylęgała
- Health Promotion and Obesity Management, Pathophysiology Department, Medical University of Silesia in Katowice, 40-752 Katowice, Poland;
| |
Collapse
|
8
|
Vu QV, Sayama S, Ando M, Kataoka T. Sesquiterpene Lactones Containing an α-Methylene-γ-Lactone Moiety Selectively Down-Regulate the Expression of Tumor Necrosis Factor Receptor 1 by Promoting Its Ectodomain Shedding in Human Lung Adenocarcinoma A549 Cells. Molecules 2024; 29:1866. [PMID: 38675685 PMCID: PMC11053566 DOI: 10.3390/molecules29081866] [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: 03/19/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Alantolactone is a eudesmane-type sesquiterpene lactone containing an α-methylene-γ-lactone moiety. Previous studies showed that alantolactone inhibits the nuclear factor κB (NF-κB) signaling pathway by targeting the inhibitor of NF-κB (IκB) kinase. However, in the present study, we demonstrated that alantolactone selectively down-regulated the expression of tumor necrosis factor (TNF) receptor 1 (TNF-R1) in human lung adenocarcinoma A549 cells. Alantolactone did not affect the expression of three adaptor proteins recruited to TNF-R1. The down-regulation of TNF-R1 expression by alantolactone was suppressed by an inhibitor of TNF-α-converting enzyme. Alantolactone increased the soluble forms of TNF-R1 that were released into the culture medium as an ectodomain. The structure-activity relationship of eight eudesmane derivatives revealed that an α-methylene-γ-lactone moiety was needed to promote TNF-R1 ectodomain shedding. In addition, parthenolide and costunolide, two sesquiterpene lactones with an α-methylene-γ-lactone moiety, increased the amount of soluble TNF-R1. Therefore, the present results demonstrate that sesquiterpene lactones with an α-methylene-γ-lactone moiety can down-regulate the expression of TNF-R1 by promoting its ectodomain shedding in A549 cells.
Collapse
Affiliation(s)
- Quy Van Vu
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Shinsei Sayama
- Department of Natural Sciences (Chemistry), Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan;
| | - Masayoshi Ando
- Department of Chemistry and Chemical Engineering, Niigata University, 2-8050 Ikarashi, Nishi-ku, Niigata 950-2181, Japan
| | - Takao Kataoka
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
- Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| |
Collapse
|
9
|
Álvarez E, Falqui M, Sin L, McGrail JP, Perdiguero B, Coloma R, Marcos-Villar L, Tárrega C, Esteban M, Gómez CE, Guerra S. Unveiling the Multifaceted Roles of ISG15: From Immunomodulation to Therapeutic Frontiers. Vaccines (Basel) 2024; 12:153. [PMID: 38400136 PMCID: PMC10891536 DOI: 10.3390/vaccines12020153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
The Interferon Stimulated Gene 15 (ISG15), a unique Ubiquitin-like (Ubl) modifier exclusive to vertebrates, plays a crucial role in the immune system. Primarily induced by interferon (IFN) type I, ISG15 functions through diverse mechanisms: (i) covalent protein modification (ISGylation); (ii) non-covalent intracellular action; and (iii) exerting extracellular cytokine activity. These various roles highlight its versatility in influencing numerous cellular pathways, encompassing DNA damage response, autophagy, antiviral response, and cancer-related processes, among others. The well-established antiviral effects of ISGylation contrast with its intriguing dual role in cancer, exhibiting both suppressive and promoting effects depending on the tumour type. The multifaceted functions of ISG15 extend beyond intracellular processes to extracellular cytokine signalling, influencing immune response, chemotaxis, and anti-tumour effects. Moreover, ISG15 emerges as a promising adjuvant in vaccine development, enhancing immune responses against viral antigens and demonstrating efficacy in cancer models. As a therapeutic target in cancer treatment, ISG15 exhibits a double-edged nature, promoting or suppressing oncogenesis depending on the tumour context. This review aims to contribute to future studies exploring the role of ISG15 in immune modulation and cancer therapy, potentially paving the way for the development of novel therapeutic interventions, vaccine development, and precision medicine.
Collapse
Affiliation(s)
- Enrique Álvarez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.S.); (B.P.); (L.M.-V.); (M.E.)
| | - Michela Falqui
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.F.); (J.P.M.); (R.C.); (C.T.)
| | - Laura Sin
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.S.); (B.P.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Joseph Patrick McGrail
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.F.); (J.P.M.); (R.C.); (C.T.)
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.S.); (B.P.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Rocío Coloma
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.F.); (J.P.M.); (R.C.); (C.T.)
| | - Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.S.); (B.P.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Céline Tárrega
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.F.); (J.P.M.); (R.C.); (C.T.)
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.S.); (B.P.); (L.M.-V.); (M.E.)
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.S.); (B.P.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Susana Guerra
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.F.); (J.P.M.); (R.C.); (C.T.)
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
10
|
Pan W, Zhang J, Zhang L, Zhang Y, Song Y, Han L, Tan M, Yin Y, Yang T, Jiang T, Li H. Comprehensive view of macrophage autophagy and its application in cardiovascular diseases. Cell Prolif 2024; 57:e13525. [PMID: 37434325 PMCID: PMC10771119 DOI: 10.1111/cpr.13525] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 07/13/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the primary drivers of the growing public health epidemic and the leading cause of premature mortality and economic burden worldwide. With decades of research, CVDs have been proven to be associated with the dysregulation of the inflammatory response, with macrophages playing imperative roles in influencing the prognosis of CVDs. Autophagy is a conserved pathway that maintains cellular functions. Emerging evidence has revealed an intrinsic connection between autophagy and macrophage functions. This review focuses on the role and underlying mechanisms of autophagy-mediated regulation of macrophage plasticity in polarization, inflammasome activation, cytokine secretion, metabolism, phagocytosis, and the number of macrophages. In addition, autophagy has been shown to connect macrophages and heart cells. It is attributed to specific substrate degradation or signalling pathway activation by autophagy-related proteins. Referring to the latest reports, applications targeting macrophage autophagy have been discussed in CVDs, such as atherosclerosis, myocardial infarction, heart failure, and myocarditis. This review describes a novel approach for future CVD therapies.
Collapse
Affiliation(s)
- Wanqian Pan
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Jun Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Lei Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yue Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yiyi Song
- Suzhou Medical College of Soochow UniversitySuzhouChina
| | - Lianhua Han
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Mingyue Tan
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yunfei Yin
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Tianke Yang
- Department of Ophthalmology, Eye Institute, Eye & ENT HospitalFudan UniversityShanghaiChina
- Department of OphthalmologyThe First Affiliated Hospital of USTC, University of Science and Technology of ChinaHefeiChina
| | - Tingbo Jiang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Hongxia Li
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
11
|
Wu M, Xu X, Hu R, Chen Q, Chen L, Yuan Y, Li J, Zhou L, Feng S, Wang L, Chen S, Gu M. A Membrane-Targeted Photosensitizer Prevents Drug Resistance and Induces Immune Response in Treating Candidiasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207736. [PMID: 37875397 PMCID: PMC10724446 DOI: 10.1002/advs.202207736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 09/20/2023] [Indexed: 10/26/2023]
Abstract
Candida albicans (C. albicans), a ubiquitous polymorphic fungus in humans, causes different types of candidiasis, including oral candidiasis (OC) and vulvovaginal candidiasis (VVC), which are physically and mentally concerning and financially costly. Thus, developing alternative antifungals that prevent drug resistance and induce immunity to eliminate Candida biofilms is crucial. Herein, a novel membrane-targeted aggregation-induced emission (AIE) photosensitizer (PS), TBTCP-QY, is developed for highly efficient photodynamic therapy (PDT) of candidiasis. TBTCP-QY has a high molar absorption coefficient and an excellent ability to generate 1 O2 and •OH, entering the interior of biofilms due to its high permeability. Furthermore, TBTCP-QY can efficiently inhibit biofilm formation by suppressing the expression of genes related to the adhesion (ALS3, EAP1, and HWP1), invasion (SAP1 and SAP2), and drug resistance (MDR1) of C. albicans, which is also advantageous for eliminating potential fungal resistance to treat clinical infectious diseases. TBTCP-QY-mediated PDT efficiently targets OC and VVC in vivo in a mouse model, induces immune response, relieves inflammation, and accelerates the healing of mucosal defects to combat infections caused by clinically isolated fluconazole-resistant strains. Moreover, TBTCP-QY demonstrates excellent biocompatibility, suggesting its potential applications in the clinical treatment of OC and VVC.
Collapse
Affiliation(s)
- Ming‐Yu Wu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural DrugsSchool of Life Science and EngineeringSouthwest Jiaotong UniversityChengduSichuan610031China
| | - Xiaoyu Xu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Rui Hu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| | - Qingrong Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Luojia Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Yuncong Yuan
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Jie Li
- Department of Medical Intensive Care UnitMaternal and Child Health Hospital of Hubei ProvinceTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430070China
| | - Li Zhou
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Shun Feng
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural DrugsSchool of Life Science and EngineeringSouthwest Jiaotong UniversityChengduSichuan610031China
| | - Lianrong Wang
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| | - Shi Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Meijia Gu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| |
Collapse
|
12
|
Halász H, Szatmári Z, Kovács K, Koppán M, Papp S, Szabó-Meleg E, Szatmári D. Changes of Ex Vivo Cervical Epithelial Cells Due to Electroporation with JMY. Int J Mol Sci 2023; 24:16863. [PMID: 38069185 PMCID: PMC10706833 DOI: 10.3390/ijms242316863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/20/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
The ionic environment within the nucleoplasm might diverge from the conditions found in the cytoplasm, potentially playing a role in the cellular stress response. As a result, it is conceivable that interactions of nuclear actin and actin-binding proteins (ABPs) with apoptosis factors may differ in the nucleoplasm and cytoplasm. The primary intracellular stress response is Ca2+ influx. The junctional mediating and regulating Y protein (JMY) is an actin-binding protein and has the capability to interact with the apoptosis factor p53 in a Ca2+-dependent manner, forming complexes that play a regulatory role in cytoskeletal remodelling and motility. JMY's presence is observed in both the cytoplasm and nucleoplasm. Here, we show that ex vivo ectocervical squamous cells subjected to electroporation with JMY protein exhibited varying morphological alterations. Specifically, the highly differentiated superficial and intermediate cells displayed reduced nuclear size. In inflamed samples, nuclear enlargement and simultaneous cytoplasmic reduction were observable and showed signs of apoptotic processes. In contrast, the less differentiated parabasal and metaplastic cells showed increased cytoplasmic activity and the formation of membrane protrusions. Surprisingly, in severe inflammation, vaginosis or ASC-US (Atypical Squamous Cells of Undetermined Significance), JMY appears to influence only the nuclear and perinuclear irregularities of differentiated cells, and cytoplasmic abnormalities still existed after the electroporation. Our observations can provide an appropriate basis for the exploration of the relationship between cytopathologically relevant morphological changes of epithelial cells and the function of ABPs. This is particularly important since ABPs are considered potential diagnostic and therapeutic biomarkers for both cancers and chronic inflammation.
Collapse
Affiliation(s)
- Henriett Halász
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (H.H.); (E.S.-M.)
| | | | - Krisztina Kovács
- Department of Pathology, Medical School, University of Pécs, 7624 Pécs, Hungary;
| | | | - Szilárd Papp
- DaVinci Clinics, 7635 Pécs, Hungary; (M.K.); (S.P.)
| | - Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (H.H.); (E.S.-M.)
| | - Dávid Szatmári
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (H.H.); (E.S.-M.)
| |
Collapse
|
13
|
Díaz-Alvarez L, Martínez-Sánchez ME, Gray E, Pérez-Figueroa E, Ortega E. Aminopeptidase N/CD13 Crosslinking Promotes the Activation and Membrane Expression of Integrin CD11b/CD18. Biomolecules 2023; 13:1488. [PMID: 37892170 PMCID: PMC10604325 DOI: 10.3390/biom13101488] [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: 08/23/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
The β2 integrin CD11b/CD18, also known as complement receptor 3 (CR3), and the moonlighting protein aminopeptidase N (CD13), are two myeloid immune receptors with overlapping activities: adhesion, migration, phagocytosis of opsonized particles, and respiratory burst induction. Given their common functions, shared physical location, and the fact that some receptors can activate a selection of integrins, we hypothesized that CD13 could induce CR3 activation through an inside-out signaling mechanism and possibly have an influence on its membrane expression. We revealed that crosslinking CD13 on the surface of human macrophages not only activates CR3 but also influences its membrane expression. Both phenomena are affected by inhibitors of Src, PLCγ, Syk, and actin polymerization. Additionally, after only 10 min at 37 °C, cells with crosslinked CD13 start secreting pro-inflammatory cytokines like interferons type 1 and 2, IL-12p70, and IL-17a. We integrated our data with a bioinformatic analysis to confirm the connection between these receptors and to suggest the signaling cascade linking them. Our findings expand the list of features of CD13 by adding the activation of a different receptor via inside-out signaling. This opens the possibility of studying the joint contribution of CD13 and CR3 in contexts where either receptor has a recognized role, such as the progression of some leukemias.
Collapse
Affiliation(s)
- Laura Díaz-Alvarez
- Instituto de Investigaciones Biomédicas, Departamento de Inmunología, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Mexico City 04510, Mexico
| | | | - Eleanor Gray
- London Centre for Nanotechnology, Department of Physics and Astronomy, University College London, London WC2R 2LS, UK
| | - Erandi Pérez-Figueroa
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| | - Enrique Ortega
- Instituto de Investigaciones Biomédicas, Departamento de Inmunología, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
14
|
Gerosa L, Malvandi AM, Malavolta M, Provinciali M, Lombardi G. Exploring cellular senescence in the musculoskeletal system: Any insights for biomarkers discovery? Ageing Res Rev 2023; 88:101943. [PMID: 37142059 DOI: 10.1016/j.arr.2023.101943] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/21/2023] [Accepted: 05/01/2023] [Indexed: 05/06/2023]
Abstract
The locomotor system comprises skeletal muscles and bones with active metabolism and cellular turnover. Chronic locomotor system disorders gradually arising with aging are inversely associated with the correct function of bone and muscles. Senescent cells appear more frequently in advanced ages or pathological conditions, and the accumulation of senescent cells in muscle tissue negatively correlates with muscle regeneration, which is crucial for maintaining strength and preventing frailty. Senescence in the bone microenvironment, osteoblasts, and osteocytes affects bone turnover favoring osteoporosis. It is likely that in response to injury and age-related damage over the lifetime, a subset of niche cells accumulates oxidative stress and DNA damage beyond the threshold that primes the onset of cellular senescence. These senescent cells may acquire resistance to apoptosis that, combined with the weakened immune system, results in impaired clearance of senescent cells and their accumulation. The secretory profile of senescent cells causes local inflammation, further spreading senescence in neighboring niche cells and impairing tissue homeostasis. The resulting impairment of turnover/tissue repair in the musculoskeletal system reduces the efficiency of the organ in response to environmental needs that finally lead to functional decline. Management of the musculoskeletal system at the cellular level can benefit the quality of life and reduce early aging. This work discusses current knowledge of cellular senescence of musculoskeletal tissues to conclude with biologically active biomarkers effective enough to reveal the underlying mechanisms of tissue flaws at the earliest possible.
Collapse
Affiliation(s)
- Laura Gerosa
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Amir Mohammad Malvandi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy.
| | - Marco Malavolta
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121 Ancona, Italy
| | - Mauro Provinciali
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121 Ancona, Italy
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy; Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| |
Collapse
|
15
|
Du X, Cui X, Sun X, Li H, Xu K, Fu X. Platycodin D-Induced Immunotoxicity in RAW 264.7 Macrophages via Oxidative Stress-Mediated Apoptosis. Nat Prod Commun 2023. [DOI: 10.1177/1934578x221150366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Platycodin D (PD) is a naturally occurring, biologically active triterpenoid saponin isolated from a medicinal food homology plant called Platycodon grandiflorus (Jacq.) A. DC. It is involved in the processing of various biological activities. While investigating the anti-inflammatory property of PD using lipopolysaccharide (LPS)-stimulated murine RAW 264.7 macrophage cells, we unexpectedly found that PD exhibited toxicity to RAW 264.7 cells. In this study, the toxic effect of PD on RAW 264.7 cells was systematically evaluated for the first time. The results showed that PD (12.5−200 µM) significantly reduced cell viability and inhibited cell proliferation in a dose-dependent manner. At a concentration of 20 µM, PD significantly increased lactate dehydrogenase activity and the mRNA and protein expression of Bax, p53, Casp3, IL-1β, and TNF-α. Interestingly, PD (0.8−20 µM) inhibited the expression of inflammatory cytokines in LPS-stimulated RAW 264.7 cells. PD (20 µM) also significantly increased reactive oxygen species (ROS) levels and the expression of oxidative stress-related genes and proteins. This study revealed that PD exhibited immunotoxicity to RAW 264.7 cells, with possible mechanisms including oxidative stress-mediated apoptosis resulting in activation of the mitochondrial apoptosis pathway and dysregulated expression of inflammatory cytokines. This study evaluated the impact of PD on immunity and provided guidelines for its future biological application.
Collapse
Affiliation(s)
- Xinying Du
- Marine Traditional Chinese Medicine Research Center, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong University of Traditional Chinese Medicine, Qingdao Academy of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine's Deep Development and Industrialization, Qingdao, China
| | - Xinhai Cui
- Marine Traditional Chinese Medicine Research Center, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaowen Sun
- Marine Traditional Chinese Medicine Research Center, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hui Li
- Marine Traditional Chinese Medicine Research Center, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong University of Traditional Chinese Medicine, Qingdao Academy of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine's Deep Development and Industrialization, Qingdao, China
| | - Kuo Xu
- Marine Traditional Chinese Medicine Research Center, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong University of Traditional Chinese Medicine, Qingdao Academy of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine's Deep Development and Industrialization, Qingdao, China
| | - Xianjun Fu
- Marine Traditional Chinese Medicine Research Center, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong University of Traditional Chinese Medicine, Qingdao Academy of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine's Deep Development and Industrialization, Qingdao, China
| |
Collapse
|
16
|
Domínguez-Horta MDC, Serrano-Díaz A, Hernández-Cedeño M, Martínez-Donato G, Guillén-Nieto G. A peptide derived from HSP60 reduces proinflammatory cytokines and soluble mediators: a therapeutic approach to inflammation. Front Immunol 2023; 14:1162739. [PMID: 37187739 PMCID: PMC10179499 DOI: 10.3389/fimmu.2023.1162739] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Cytokines are secretion proteins that mediate and regulate immunity and inflammation. They are crucial in the progress of acute inflammatory diseases and autoimmunity. In fact, the inhibition of proinflammatory cytokines has been widely tested in the treatment of rheumatoid arthritis (RA). Some of these inhibitors have been used in the treatment of COVID-19 patients to improve survival rates. However, controlling the extent of inflammation with cytokine inhibitors is still a challenge because these molecules are redundant and pleiotropic. Here we review a novel therapeutic approach based on the use of the HSP60-derived Altered Peptide Ligand (APL) designed for RA and repositioned for the treatment of COVID-19 patients with hyperinflammation. HSP60 is a molecular chaperone found in all cells. It is involved in a wide diversity of cellular events including protein folding and trafficking. HSP60 concentration increases during cellular stress, for example inflammation. This protein has a dual role in immunity. Some HSP60-derived soluble epitopes induce inflammation, while others are immunoregulatory. Our HSP60-derived APL decreases the concentration of cytokines and induces the increase of FOXP3+ regulatory T cells (Treg) in various experimental systems. Furthermore, it decreases several cytokines and soluble mediators that are raised in RA, as well as decreases the excessive inflammatory response induced by SARS-CoV-2. This approach can be extended to other inflammatory diseases.
Collapse
Affiliation(s)
- Maria del Carmen Domínguez-Horta
- Autoimmunity Project, Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
- Physiology Department, Latin American School of Medicine, Havana, Cuba
- *Correspondence: Maria del Carmen Domínguez-Horta,
| | - Anabel Serrano-Díaz
- Autoimmunity Project, Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Mabel Hernández-Cedeño
- Autoimmunity Project, Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Gillian Martínez-Donato
- Biomedical Research Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Gerardo Guillén-Nieto
- Physiology Department, Latin American School of Medicine, Havana, Cuba
- Biomedical Research Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| |
Collapse
|
17
|
Bove GM, Mokler DJ. Effects of a single dose of psilocybin on cytokines, chemokines and leptin in rat serum. JOURNAL OF PSYCHEDELIC STUDIES 2022. [DOI: 10.1556/2054.2022.00230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AbstractBackground and AimsThe hallucinogenic drug psilocybin is being widely tested in humans for the treatment of psychiatric disorders. Psilocybin and other psychedelics are proposed to work through serotonin 2a (5-HT2a) receptors, which are tightly linked to immune function. The purpose of the present study was to assess the effects of a single dose of psilocybin on a panel of cytokines, chemokines, and peptides in the short term (24 h) and long term (seven days) in female rats.MethodsFemale rats were given a dose of psilocybin (20 mg kg−1, i.p.} or a dose of synthetic interstitial fluid. At 24 h, the control group and one group of rats were anesthetized, and blood was withdrawn by intracardiac puncture. In a third group of rats, blood was withdrawn after seven days. Serum was analyzed by a separate lab (Eve Laboratories, Calgary, Canada) for 27 immunomodulators.ResultsSerum levels of IL-1β, TNF-α, MCP-1, IP-10, G-CSF, IFN-γ, IL-10, IL-13, and leptin were significantly increased compared to controls after 24 h and were increased further after 7 days. Most of the other assays showed this same pattern of increase, although not statistically significant.ConclusionsPsilocybin induces the release of multiple immune factors, consistent with a generalized activation of the immune system, which can persist for at least seven days after a single dose. These findings may relate to the mechanism of action. The implications of these findings require additional research to determine how these finding relate to the clinical effects of psilocybin.
Collapse
|
18
|
Zhao H, Zhang J, Rajeshkumar S, Feng Y, Liu Y, Li X, Zhang B. Hepatopancreas toxicity and immunotoxicity of a fungicide, pyraclostrobin, on common carp. Comp Biochem Physiol C Toxicol Pharmacol 2022; 262:109445. [PMID: 36030005 DOI: 10.1016/j.cbpc.2022.109445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/31/2022] [Accepted: 08/21/2022] [Indexed: 11/23/2022]
Abstract
Pyraclostrobin (PYR), a strobilurin fungicide, has been widely used to control fungal diseases, posing potential risk to aquatic organisms. However, the toxic effects of PYR to fish remained largely unknown. In this study, common carp (Cyprinus carpio L.) was exposed to environmentally relevant levels of PYR (0, 0.5 and 5.0 μg/L) for 30 days to assess its chronic toxicity and potential toxicity mechanism. The results showed that long-term exposure to PYR induced hepatopancreas damage as evident by increased in serum transaminase activities (AST and ALT). Moreover, PYR exposure remarkably enhanced the expressions of hsp70 and hsp90, decreased the levels of antioxidant enzymes and biomarkers and promoted the reactive oxygen species (H2O2 and O2-) and MDA contents in carp hepatopancreas. PYR exposure also upregulated apoptosis-related genes (bax, apaf-1, caspase-3 and caspase-9) and reduced anti-apoptosis gene bcl-2 in fish hepatopancreas. Moreover, PYR exposure altered the expressions of inflammatory cytokines (IL-1β, IL-6, TNF-α and TGF-β) in the serum and hepatopancreas and the level of NF-κB p65 in the hepatopancreas. Further research indicated that PYR exposure markedly changed the levels of immune parameters (LYZ, C3, IgM, ACP and AKP) in the serum and/or hepatopancreas, indicating that chronic PYR exposure also has immunotoxicity on fish. Additionally, we found that PYR exposure upregulated p38 and jnk MAPK transcription levels, suggesting that MAPK may be play important role in PYR-induced apoptosis and inflammatory response in the hepatopancreas of common carp. In summary, PYR exposure induced oxidative stress, triggered apoptosis, inflammatory and immune response in common carp, which can help to elucidate the possible toxicity mechanism of PYR in fish.
Collapse
Affiliation(s)
- Haoyang Zhao
- Henan International Joint Laboratory of Aquatic Ecotoxicology and Health Protection, College of Life Sciences, Henan Normal University, Xinxiang 453007, China
| | - Jiale Zhang
- Henan International Joint Laboratory of Aquatic Ecotoxicology and Health Protection, College of Life Sciences, Henan Normal University, Xinxiang 453007, China
| | | | - Yiyi Feng
- Henan International Joint Laboratory of Aquatic Ecotoxicology and Health Protection, College of Life Sciences, Henan Normal University, Xinxiang 453007, China
| | - Yang Liu
- Henan International Joint Laboratory of Aquatic Ecotoxicology and Health Protection, College of Life Sciences, Henan Normal University, Xinxiang 453007, China; Journal of Henan Normal University, Xinxiang 453007, China
| | - Xiaoyu Li
- Henan International Joint Laboratory of Aquatic Ecotoxicology and Health Protection, College of Life Sciences, Henan Normal University, Xinxiang 453007, China
| | - Bangjun Zhang
- Henan International Joint Laboratory of Aquatic Ecotoxicology and Health Protection, College of Life Sciences, Henan Normal University, Xinxiang 453007, China.
| |
Collapse
|
19
|
Kumar N, Vyas A, Agnihotri SK, Chattopadhyay N, Sachdev M. Small secretory proteins of immune cells can modulate gynecological cancers. Semin Cancer Biol 2022; 86:513-531. [PMID: 35150864 DOI: 10.1016/j.semcancer.2022.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 01/27/2023]
Abstract
Small secretory proteins of immune cells are mostly Cytokines, which include chemokines, interleukins, interferons, lymphokines and tumor necrosis factors but not hormones or growth factors. These secretory proteins are the molecular messengers and primarily involved in autocrine, paracrine and endocrine signaling as immunomodulating agents. Hence, these proteins actually regulate the cells of immune system to communicate with one another to produce a synchronized, robust, still self-regulated response to a specific antigen. Chemokines are smaller secreted proteins that control overall immune cell movement and location; these chemokines are divided into 4 subgroups, namely, CXC, CC, CX3C and C according to the position of 4 conserved cysteine residues. Complete characterization of cytokines and chemokines can exploit their vast signaling networks to develop cancer treatments. These secretory proteins like IL-6, IL-10, IL-12, TNFα, CCL2, CXCL4 & CXCL8 are predominantly expressed in most of the gynecological cancers, which directly stimulate immune effector cells and stromal cells at the tumor site and augment tumor cell recognition by cytotoxic T-cells. Hence; these secretory proteins are the major regulators, which can actually modulate all kinds of gynecological cancers. Furthermore, advancements in adoptive T-cell treatment have relied on the use of multiple cytokines/chemokines to establish a highly regulated environment for anti-tumor T cell growth. A number of in vitro studies as well as animal models and clinical subjects have also shown that cytokines/chemokines have broad antitumor activity, which has been translated into a number of cancer therapy approaches. This review will focus on the foremost cytokines & chemokines involved in the majority of the gynecological malignancies and discuss their basic biology as well as clinical applications.
Collapse
Affiliation(s)
- Niranjan Kumar
- Division of Endocrinology, CSIR- Central Drug Research Institute, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, 201 002, India
| | - Akanksha Vyas
- Division of Endocrinology, CSIR- Central Drug Research Institute, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, 201 002, India
| | | | - Naibedya Chattopadhyay
- Division of Endocrinology, CSIR- Central Drug Research Institute, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, 201 002, India.
| | - Monika Sachdev
- Division of Endocrinology, CSIR- Central Drug Research Institute, Lucknow, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, 201 002, India.
| |
Collapse
|
20
|
Fernandez A, Kielland N, Makda A, Carragher NO, González-García MC, Espinar-Barranco L, González-Vera JA, Orte A, Lavilla R, Vendrell M. A multicomponent reaction platform towards multimodal near-infrared BODIPY dyes for STED and fluorescence lifetime imaging. RSC Chem Biol 2022; 3:1251-1259. [PMID: 36320886 PMCID: PMC9533399 DOI: 10.1039/d2cb00168c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/25/2022] [Indexed: 11/07/2023] Open
Abstract
We report a platform combining multicomponent reaction synthesis and automated cell-based screening to develop biocompatible NIR-BODIPY fluorophores. From a library of over 60 fluorophores, we optimised compound NIRBD-62c as a multimodal probe with suitable properties for STED super-resolution and fluorescence lifetime imaging. Furthermore, we employed NIRBD-62c for imaging trafficking inside cells and to examine how pharmacological inhibitors can alter the vesicular traffic between intracellular compartments and the plasma membrane.
Collapse
Affiliation(s)
- Antonio Fernandez
- Centre for Inflammation Research, The University of Edinburgh Edinburgh UK
- Dpt Organic Chemistry, Faculty of Chemistry, University of Murcia Spain
| | - Nicola Kielland
- Centre for Inflammation Research, The University of Edinburgh Edinburgh UK
- Laboratory of Medicinal Chemistry, Faculty of Pharmacy and Institute of Biomedicine (IBUB), University of Barcelona Spain
| | - Ashraff Makda
- Institute of Genetics and Cancer, The University of Edinburgh Edinburgh UK
| | - Neil O Carragher
- Institute of Genetics and Cancer, The University of Edinburgh Edinburgh UK
| | | | | | - Juan A González-Vera
- Nanoscopy-UGR Laboratory, Facultad de Farmacia, Universidad de Granada Granada Spain
| | - Angel Orte
- Nanoscopy-UGR Laboratory, Facultad de Farmacia, Universidad de Granada Granada Spain
| | - Rodolfo Lavilla
- Laboratory of Medicinal Chemistry, Faculty of Pharmacy and Institute of Biomedicine (IBUB), University of Barcelona Spain
| | - Marc Vendrell
- Centre for Inflammation Research, The University of Edinburgh Edinburgh UK
| |
Collapse
|
21
|
An C, Wu Y, Wu J, Liu H, Zhou S, Ge D, Dong R, You L, Hao Y. Berberine ameliorates pulmonary inflammation in mice with influenza viral pneumonia by inhibiting NLRP3 inflammasome activation and gasdermin D‐mediated pyroptosis. Drug Dev Res 2022; 83:1707-1721. [DOI: 10.1002/ddr.21995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/10/2022] [Accepted: 08/23/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Chen An
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Yanmin Wu
- Department of Immunology, School of Medical Technology Qiqihar Medical University Qiqihar China
| | - Jun Wu
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Huanwei Liu
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Siyao Zhou
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Dongyu Ge
- Research and Test Center, School of Traditional Chinese Medicine Beijing University of Chinese Medicine Beijing China
| | - Ruijuan Dong
- Research and Test Center, School of Traditional Chinese Medicine Beijing University of Chinese Medicine Beijing China
| | - Leiming You
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| | - Yu Hao
- Department of Immunology and Microbiology, School of Life Science Beijing University of Chinese Medicine Beijing China
| |
Collapse
|
22
|
Biswal MR, Padmanabhan S, Manjithaya R, Prakash MK. Early Bioinformatic Implication of Triacidic Amino Acid Motifs in Autophagy-Dependent Unconventional Secretion of Mammalian Proteins. Front Cell Dev Biol 2022; 10:863825. [PMID: 35646924 PMCID: PMC9136135 DOI: 10.3389/fcell.2022.863825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/11/2022] [Indexed: 11/22/2022] Open
Abstract
Several proteins are secreted outside the cell, and in many cases, they may be identified by a characteristic signal peptide. However, more and more studies point to the evidence for an “unconventional” secretion, where proteins without a hitherto unknown signal are secreted, possibly in conditions of starvation. In this work, we analyse a set of 202 RNA binding mammalian proteins, whose unconventional secretion has recently been established. Analysis of these proteins secreted by LC3 mediation, the largest unconventionally secreted dataset to our knowledge, identifies the role of KKX motif as well as triacidic amino acid motif in unconventional secretion, the latter being an extension of the recent implicated diacidic amino acid motif. Further data analysis evolves a hypothesis on the sequence or structural proximity of the triacidic or KKX motifs to the LC3 interacting region, and a phosphorylatable amino acid such as serine as a statistically significant feature among these unconventionally secreted proteins. This hypothesis, although needs to be validated in experiments that challenge the specific details of each of these aspects, appears to be one of the early steps in defining what may be a plausible signal for unconventional protein secretion.
Collapse
Affiliation(s)
- Malay Ranjan Biswal
- Computational Biology, Theoretical Sciences Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, India
| | - Sreedevi Padmanabhan
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, India
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, India
- *Correspondence: Ravi Manjithaya, ; Meher K. Prakash,
| | - Meher K. Prakash
- Computational Biology, Theoretical Sciences Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, India
- *Correspondence: Ravi Manjithaya, ; Meher K. Prakash,
| |
Collapse
|
23
|
Bajaj R, Warner AN, Fradette JF, Gibbons DL. Dance of The Golgi: Understanding Golgi Dynamics in Cancer Metastasis. Cells 2022; 11:1484. [PMID: 35563790 PMCID: PMC9102947 DOI: 10.3390/cells11091484] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 12/17/2022] Open
Abstract
The Golgi apparatus is at the center of protein processing and trafficking in normal cells. Under pathological conditions, such as in cancer, aberrant Golgi dynamics alter the tumor microenvironment and the immune landscape, which enhances the invasive and metastatic potential of cancer cells. Among these changes in the Golgi in cancer include altered Golgi orientation and morphology that contribute to atypical Golgi function in protein trafficking, post-translational modification, and exocytosis. Golgi-associated gene mutations are ubiquitous across most cancers and are responsible for modifying Golgi function to become pro-metastatic. The pharmacological targeting of the Golgi or its associated genes has been difficult in the clinic; thus, studying the Golgi and its role in cancer is critical to developing novel therapeutic agents that limit cancer progression and metastasis. In this review, we aim to discuss how disrupted Golgi function in cancer cells promotes invasion and metastasis.
Collapse
Affiliation(s)
- Rakhee Bajaj
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Amanda N. Warner
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Jared F. Fradette
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
| | - Don L. Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
24
|
Yang PS, Yu MH, Hou YC, Chang CP, Lin SC, Kuo IY, Su PC, Cheng HC, Su WC, Shan YS, Wang YC. Targeting protumor factor chitinase-3-like-1 secreted by Rab37 vesicles for cancer immunotherapy. Am J Cancer Res 2022; 12:340-361. [PMID: 34987649 PMCID: PMC8690922 DOI: 10.7150/thno.65522] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Chitinase 3-like-1 (CHI3L1) is a secretion glycoprotein associated with the immunosuppressive tumor microenvironment (TME). The secretory mode of CHI3L1 makes it a promising target for cancer treatment. We have previously reported that Rab37 small GTPase mediates secretion of IL-6 in macrophages to promote cancer progression, whereas the roles of Rab37 in the intracellular trafficking and exocytosis of CHI3L1 are unclear. Methods: We examined the concentration of CHI3L1 in the culture medium of splenocytes and bone marrow derived macrophages (BMDMs) from wild-type or Rab37 knockout mice, and macrophage or T cell lines expressing wild type, active GTP-bound or inactive GDP-bound Rab37. Vesicle isolation, total internal reflection fluorescence microscopy, and real-time confocal microscopy were conducted. We developed polyclonal neutralizing-CHI3L1 antibodies (nCHI3L1 Abs) to validate the therapeutic efficacy in orthotopic lung, pancreas and colon cancer allograft models. Multiplex fluorescence immunohistochemistry was performed to detect the protein level of Rab37 and CHI3L1, and localization of the tumor-infiltrating immune cells in allografts from mice or tumor specimens from cancer patients. Results: We demonstrate a novel secretion mode of CHI3L1 mediated by the small GTPase Rab37 in T cells and macrophages. Rab37 mediated CHI3L1 intracellular vesicle trafficking and exocytosis in a GTP-dependent manner, which is abolished in the splenocytes and BMDMs from Rab37 knockout mice and attenuated in macrophage or T cell lines expressing the inactive Rab37. The secreted CHI3L1 activated AKT, ß-catenin and NF-κB signal pathways in cancer cells and macrophages to foster a protumor TME characterized by activating M2 macrophages and increasing the population of regulatory T cells. Our developed nCHI3L1 Abs showed the dual properties of reducing tumor growth/metastases and eliciting an immunostimulatory TME in syngeneic orthotopic lung, pancreas and colon tumor models. Clinically, high plasma level or intratumoral expression of CHI3L1 correlated with poor survival in 161 lung cancer, 155 pancreatic cancer and 180 colon cancer patients. Conclusions: These results provide the first evidence that Rab37 mediates CHI3L1 secretion in immune cells and highlight nCHI3L1 Abs that can simultaneously target both cancer cells and tumor microenvironment.
Collapse
|
25
|
Cordeiro PAS, Assone T, Prates G, Tedeschi MRM, Fonseca LAM, Casseb J. The role of IFN-γ production during retroviral infections: an important cytokine involved in chronic inflammation and pathogenesis. Rev Inst Med Trop Sao Paulo 2022; 64:e64. [PMID: 36197425 PMCID: PMC9528752 DOI: 10.1590/s1678-9946202264064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
Interferon-gamma (IFN-γ) plays a crucial role in viral infections by preventing viral replication and in the promotion of innate and adaptive immune responses. However, IFN-gamma can exert distinct effects in different persistent viral infections. The long-term overproduction of IFN-γ in retroviral infections, such as the human immunodeficiency virus (HIV), human T-lymphotropic virus type 1 (HTLV-1), and human endogenous retroviruses (HERVs), resulting in inflammation, may cause neuronal damage. This review is provocative about the role of IFN-γ during persistent retroviral infections and its relationship with the causation of some neurological disorders that are important for public health.
Collapse
|
26
|
Al-Hariri M, Alsunni A, Shaikh MH, Gamal Eldin T, Al Ghamdi K, Alharbi AF, Alhawaj H, Chathoth S. Caffeic Acid Phenethyl Ester reduces Pro Inflammatory Cytokines in Moderate Swimming Test in Growing Rats Model. J Inflamm Res 2021; 14:5653-5657. [PMID: 34754212 PMCID: PMC8570724 DOI: 10.2147/jir.s338973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 10/22/2021] [Indexed: 01/13/2023] Open
Abstract
Background Caffeic acid phenethyl ester (CAPE) is a naturally occurring polyphenolic concentrated in propolis of honeybee hives. CAPE has been shown various physiological and pharmacologic properties. The aim of the present study was to investigate the effects of CAPE on proinflammatory markers in growing rats by performing the moderate swimming test. Methods A total number of 21 male Wistar albino rats were separated into three groups (n = 7): sedentary: negative control group; exercise: positive control group received vehicle orally and exercise + CAPE: CAPE treated group: treated with CAPE (20 mg/kg) orally 30 min before exercise, for 5 days. The animals were left free to swim in the tank, 20 minutes/day for 5 days. At 24 hours after finishing the experiment, rats were euthanised and blood was collected to analyze the level of serum interleukin IL-6 and tumor necrosis factor-α (TNF-α). Results Growing rats subjected to the moderate swimming test and in those treated with CAPE showed a lower level of TNF-α compared to the negative control. More interestingly, the one-way ANOVA data demonstrated a decreased level of proinflammatory IL-6 in the CAPE-treated group compared to the negative control. Conclusion Results of this study indicate that short-term administration of CAPE may modulate proinflammatory cytokine profiles during moderate exercise and may serve to boost the anti-inflammatory effects of exercise. Further studies are needed to evaluate the efficacy and safety of long-term administration of CAPE as an adjective anti-inflammatory agent.
Collapse
Affiliation(s)
- Mohammed Al-Hariri
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Ahmed Alsunni
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohammad Habeeb Shaikh
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Tharwat Gamal Eldin
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Kholoud Al Ghamdi
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Abdulelah Fawzi Alharbi
- Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Hussain Alhawaj
- Department of Environmental Health, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabi
| | - Shahanas Chathoth
- Department of Biochemistry, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
27
|
SenGupta S, Hein LE, Parent CA. The Recruitment of Neutrophils to the Tumor Microenvironment Is Regulated by Multiple Mediators. Front Immunol 2021; 12:734188. [PMID: 34567000 PMCID: PMC8461236 DOI: 10.3389/fimmu.2021.734188] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022] Open
Abstract
Neutrophils sense and migrate towards chemotactic factors released at sites of infection/inflammation and contain the affected area using a variety of effector mechanisms. Aside from these established immune defense functions, neutrophils are emerging as one of the key tumor-infiltrating immune cells that influence cancer progression and metastasis. Neutrophil recruitment to the tumor microenvironment (TME) is mediated by multiple mediators including cytokines, chemokines, lipids, and growth factors that are secreted from cancer cells and cancer-associated stromal cells. However, the molecular mechanisms that underlie the expression and secretion of the different mediators from cancer cells and how neutrophils integrate these signals to reach and invade tumors remain unclear. Here, we discuss the possible role of the epithelial to mesenchymal transition (EMT) program, which is a well-established promoter of malignant potential in cancer, in regulating the expression and secretion of these key mediators. We also summarize and review our current understanding of the machineries that potentially control the secretion of the mediators from cancer cells, including the exocytic trafficking pathways, secretory autophagy, and extracellular vesicle-mediated secretion. We further reflect on possible mechanisms by which different mediators collaborate by integrating their signaling network, and particularly focus on TGF-β, a cytokine that is highly expressed in invasive tumors, and CXCR2 ligands, which are crucial neutrophil recruiting chemokines. Finally, we highlight gaps in the field and the need to expand current knowledge of the secretory machineries and cross-talks among mediators to develop novel neutrophil targeting strategies as effective therapeutic options in the treatment of cancer.
Collapse
Affiliation(s)
- Shuvasree SenGupta
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Lauren E. Hein
- Cancer Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Carole A. Parent
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
28
|
Kuo WT, Chang JM, Chen CC, Tsao N, Chang CP. Autophagy drives plasticity and functional polarization of tumor-associated macrophages. IUBMB Life 2021; 74:157-169. [PMID: 34467634 DOI: 10.1002/iub.2543] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/23/2021] [Accepted: 08/07/2021] [Indexed: 01/11/2023]
Abstract
Tumor-associated macrophages (TAMs) are a major component of the tumor microenvironment (TME) and are key cells in regulating tumor development, metastasis, immune responses, inflammation, and chemoresistance. In response to TME stimulation, circulating monocytes are recruited and differentiated as TAMs. Most TAMs are defined as alternatively activated (M2) phenotype to create immunosuppressive TME and support tumor progression. In contrast, classically activated (M1) TAMs can produce pro-inflammatory cytokines and enhance immune responses against tumor development. Autophagy is a conserved catabolic process to control cellular homeostasis and biological function. Emerging evidence reveals crucial contribution of autophagy in modulating TAM plasticity and functional polarization in TME. In this review, we introduce the current understanding of autophagy-regulated TAM function in development of cancer. We focus on how autophagy modulates antigen presentation, LC3-associated phagocytosis, cytokine secretion, inflammasome regulation, recruitment, differentiation, and polarization of TAMs and suggest strategies for potential therapeutics by targeting autophagy in TAMs. We expect this review can provide a new notion of future cancer immunotherapy.
Collapse
Affiliation(s)
- Wan-Ting Kuo
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jia-Ming Chang
- Department of Surgery, Division of Thoracic Surgery, Chia-Yi Christian Hospital, Chiayi, Taiwan.,Department of Physical Therapy, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chien-Chin Chen
- Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan, Taiwan.,Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Nina Tsao
- Department of Medical Laboratory Science, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Chih-Peng Chang
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Microbiology & Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
29
|
Kuo IY, Yang YE, Yang PS, Tsai YJ, Tzeng HT, Cheng HC, Kuo WT, Su WC, Chang CP, Wang YC. Converged Rab37/IL-6 trafficking and STAT3/PD-1 transcription axes elicit an immunosuppressive lung tumor microenvironment. Am J Cancer Res 2021; 11:7029-7044. [PMID: 34093869 PMCID: PMC8171097 DOI: 10.7150/thno.60040] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/04/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Increased IL-6 level, M2 macrophages and PD-1+CD8+ T cells in tumor microenvironments (TME) have been identified to correlate with resistance to checkpoint blockade immunotherapy, yet the mechanism remains poorly understood. Rab small GTPase-mediated trafficking of cytokines is critical in immuno-modulation. We have previously reported dysregulation of Rab37 in lung cancer cells, whereas the roles of Rab37 in tumor-infiltrating immune cells and cancer immunotherapy are unclear. Methods: The tumor growth of the syngeneic mouse allograft in wild type or Rab37 knockout mice was analyzed. Imaging analyses and vesicle isolation were conducted to determine Rab37-mediated IL-6 secretion. STAT3 binding sites at PD-1 promoter in T cells were identified by chromatin immunoprecipitation assay. Multiplex fluorescence immunohistochemistry was performed to detect the protein level of Rab37, IL-6 and PD-1 and localization of the tumor-infiltrating immune cells in allografts from mice or tumor specimens from lung cancer patients. Results: We revealed that Rab37 regulates the secretion of IL-6 in a GTPase-dependent manner in macrophages to trigger M2 polarization. Macrophage-derived IL-6 promotes STAT3-dependent PD-1 mRNA expression in CD8+ T cells. Clinically, tumors with high stromal Rab37 and IL-6 expression coincide with tumor infiltrating M2-macrophages and PD1+CD8+ T cells that predicts poor prognosis in lung cancer patients. In addition, lung cancer patients with an increase in plasma IL-6 level are found to be associated with immunotherapeutic resistance. Importantly, combined blockade of IL-6 and CTLA-4 improves survival of tumor-bearing mice by reducing infiltration of PD1+CD8+ T cells and M2 macrophages in TME. Conclusions: Rab37/IL-6 trafficking pathway links with IL-6/STAT3/PD-1 transcription regulation to foster an immunosuppressive TME and combined IL-6/CTLA-4 blockade therapy exerts potent anti-tumor efficacy.
Collapse
|
30
|
Diaz A, Muñoz-Arenas G, Venegas B, Vázquez-Roque R, Flores G, Guevara J, Gonzalez-Vergara E, Treviño S. Metforminium Decavanadate (MetfDeca) Treatment Ameliorates Hippocampal Neurodegeneration and Recognition Memory in a Metabolic Syndrome Model. Neurochem Res 2021; 46:1151-1165. [PMID: 33559829 DOI: 10.1007/s11064-021-03250-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/02/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
The consumption of foods rich in carbohydrates, saturated fat, and sodium, accompanied by a sedentary routine, are factors that contribute to the progress of metabolic syndrome (MS). In this way, they cause the accumulation of body fat, hypertension, dyslipidemia, and hyperglycemia. Additionally, MS has been shown to cause oxidative stress, inflammation, and death of neurons in the hippocampus. Consequently, spatial and recognition memory is affected. It has recently been proposed that metformin decavanadate (MetfDeca) exerts insulin mimetic effects that enhance metabolism in MS animals; however, what effects it can cause on the hippocampal neurons of rats with MS are unknown. The objective of the work was to evaluate the effect of MetfDeca on hippocampal neurodegeneration and recognition memory in rats with MS. Administration of MetfDeca for 60 days in MS rats improved object recognition memory (NORt). In addition, MetfDeca reduced markers of oxidative stress and hippocampal neuroinflammation. Accompanied by an increase in the density and length of the dendritic spines of the hippocampus of rats with MS. We conclude that MetfDeca represents an important therapeutic agent to treat MS and induce neuronal and cognitive restoration mechanisms.
Collapse
Affiliation(s)
- Alfonso Diaz
- Faculty of Chemical Sciences, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Guadalupe Muñoz-Arenas
- Faculty of Chemical Sciences, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Berenice Venegas
- Faculty of Biological Sciences, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Rubén Vázquez-Roque
- Laboratory of Neuropsychiatry, Institute of Physiology, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Gonzalo Flores
- Laboratory of Neuropsychiatry, Institute of Physiology, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico
| | - Jorge Guevara
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | | | - Samuel Treviño
- Faculty of Chemical Sciences, Benemerita Autonomous University of Puebla, Puebla, Pue, Mexico.
| |
Collapse
|
31
|
Fathi E, Farahzadi R, Valipour B. Alginate/gelatin encapsulation promotes NK cells differentiation potential of bone marrow resident C-kit + hematopoietic stem cells. Int J Biol Macromol 2021; 177:317-327. [PMID: 33621568 DOI: 10.1016/j.ijbiomac.2021.02.131] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/16/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023]
Abstract
The ability of natural killer (NK) cells to destroy cancerous cells with no prior sensitization has made them attractive candidates for cell therapy. The application of hydrogels must be notified as cell delivery vehicles in cell differentiation. The present study was conducted to investigate the effect of alginate-gelatin encapsulation on NK cell differentiation potential of C-kit+ cells. C-kit+ cells were differentiated to NK cells under both encapsulated and un-encapsulated conditions. Next, the cells were subjected to real-time polymerase chain reaction (PCR) and western blotting for the assessment of their telomere length and protein expressions, respectively. Afterward, culture medium was collected to measure cytokines levels. Thereafter, the differentiated NK cells were co-cultured with Molt-4 cells to investigate the potency of cell apoptosis by Annexin V/PI assay. A significant change was observed in the protein expression of Janus kinase/Signal transducers (JAK/STAT) pathway components. Additionally, the encapsulation caused an increase in the apoptosis of Molt-4 cells and telomere length of NK cells differentiated C-kit+ cells. Therefore, it can be concluded that the effects of encapsulation on NK cell's differentiation of C-kit+ cells could be resulted from the secreted cytokines of interleukin (IL)-2, IL-3, IL-7, and IL-12 as well as the increased telomere length.
Collapse
Affiliation(s)
- Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Behnaz Valipour
- Department of Histopathology and Anatomy, Faculty of Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| |
Collapse
|
32
|
Kumari R, Jat P. Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype. Front Cell Dev Biol 2021; 9:645593. [PMID: 33855023 PMCID: PMC8039141 DOI: 10.3389/fcell.2021.645593] [Citation(s) in RCA: 826] [Impact Index Per Article: 206.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/16/2021] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a stable cell cycle arrest that can be triggered in normal cells in response to various intrinsic and extrinsic stimuli, as well as developmental signals. Senescence is considered to be a highly dynamic, multi-step process, during which the properties of senescent cells continuously evolve and diversify in a context dependent manner. It is associated with multiple cellular and molecular changes and distinct phenotypic alterations, including a stable proliferation arrest unresponsive to mitogenic stimuli. Senescent cells remain viable, have alterations in metabolic activity and undergo dramatic changes in gene expression and develop a complex senescence-associated secretory phenotype. Cellular senescence can compromise tissue repair and regeneration, thereby contributing toward aging. Removal of senescent cells can attenuate age-related tissue dysfunction and extend health span. Senescence can also act as a potent anti-tumor mechanism, by preventing proliferation of potentially cancerous cells. It is a cellular program which acts as a double-edged sword, with both beneficial and detrimental effects on the health of the organism, and considered to be an example of evolutionary antagonistic pleiotropy. Activation of the p53/p21WAF1/CIP1 and p16INK4A/pRB tumor suppressor pathways play a central role in regulating senescence. Several other pathways have recently been implicated in mediating senescence and the senescent phenotype. Herein we review the molecular mechanisms that underlie cellular senescence and the senescence associated growth arrest with a particular focus on why cells stop dividing, the stability of the growth arrest, the hypersecretory phenotype and how the different pathways are all integrated.
Collapse
Affiliation(s)
- Ruchi Kumari
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
| | - Parmjit Jat
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
| |
Collapse
|
33
|
Antony R, Li Y. BDNF secretion from C2C12 cells is enhanced by methionine restriction. Biochem Biophys Res Commun 2020; 533:1347-1351. [PMID: 33069357 DOI: 10.1016/j.bbrc.2020.10.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 10/08/2020] [Indexed: 11/24/2022]
Abstract
Brain derived neurotrophic factor (BDNF) is produced in skeletal muscle as a myokine that plays a role in muscle metabolism. However, how metabolic changes affect skeletal muscle BDNF expression and release remains to be fully understood. Amino acid restrictions such as methionine restriction (MR) are considered as an alternative fasting approach. Here we reported that in C2C12 myotubes, MR enhanced BDNF release, which was measured using ELISA, RT-qPCR, cell immunostaining, and Western blot. Inhibition of protein transport pathway blocked the MR enhanced BDNF release, confirming that MR-induced BDNF release involved classic protein secretory pathway. MR increased l-lactate product in media, suggesting that MR promoted glycolysis. Treatment with 2-deoxy glucose (2-DG) attenuated lactate production as well as BDNF release, suggesting that glycolysis is involved in the enhanced BDNF release induced by MR. Moreover, treatment with l-Lactate, the end-product of glycolysis, enhanced BDNF gene expression and release in control cells in a dose dependent manner, suggesting lactate produced by glycolysis may mediate the enhanced BDNF release by MR. Overall, the results of this study suggest that MR promotes BDNF secretion from C2C12 myotubes at least partially via enhancing glycolysis and lactate production.
Collapse
Affiliation(s)
- Ryan Antony
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Yifan Li
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA.
| |
Collapse
|
34
|
Rodenkirchen V, Schettgen T, Rink L. Zinc deficiency impairs interferon-γ production on post-transcriptional level. J Trace Elem Med Biol 2020; 62:126598. [PMID: 32645654 DOI: 10.1016/j.jtemb.2020.126598] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Zinc is a trace element and is thus commonly known to be indispensable for regular cellular function. Until today, zinc deficiency is a widespread health problem, affecting approximately one sixth of the world's population. Especially the immune system has proven to be highly dependent on zinc. Interferon-γ (IFN-γ) is a key element in the defense against intracellular pathogens. A lack of this cytokine results in immunological impairment, whereas an excess can lead to autoimmunity, highlighting the importance of a well-regulated IFN-γ expression. In a state of zinc deficiency, the production of this cytokine has long been shown to be reduced. Providing further insight into the molecular mechanisms responsible for this interaction is the primary objective of this study. METHODS Zinc-deficient or -supplemented cell culture, ELISA, quantitative PCR, methylation analysis. RESULTS Promoter methylation is a typical mechanism of gene silencing and a strong regulating factor for IFN-γ production. An analysis of the methylation status of IFN-γ and its transcription factor IRF4 in human PBMC in a state of cellular zinc deficiency or excess showed no dependency on the trace metal. Unexpectedly, zinc-deficient PBMC, which secreted significantly less IFN-γ protein, showed significantly higher mRNA levels of the cytokine compared to cells with high total zinc levels. CONCLUSION This report is the first about this unconventional ratio of IFN-γ mRNA to protein. Such a mismatch is highly relevant to the study of protein production in general and that of IFN-γ in particular. Based on our results and the latest research, we hypothesize a strong post-transcriptional effect of zinc on IFN-γ.
Collapse
Affiliation(s)
- Vera Rodenkirchen
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Thomas Schettgen
- Institute for Occupational, Social and Environmental Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, D-52074 Aachen, Germany
| | - Lothar Rink
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, D-52074 Aachen, Germany.
| |
Collapse
|
35
|
Karwelat D, Schmeck B, Ringel M, Benedikter BJ, Hübner K, Beinborn I, Maisner A, Schulte LN, Vollmeister E. Influenza virus-mediated suppression of bronchial Chitinase-3-like 1 secretion promotes secondary pneumococcal infection. FASEB J 2020; 34:16432-16448. [PMID: 33095949 DOI: 10.1096/fj.201902988rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022]
Abstract
Infections of the lung are among the leading causes of death worldwide. Despite the preactivation of innate defense programs during viral infection, secondary bacterial infection substantially elevates morbidity and mortality rates. Particularly problematic are co-infections with influenza A virus (IAV) and the major bacterial pathogen Streptococcus pneumoniae. However, the molecular processes underlying the severe course of such co-infections are not fully understood. Previously, the absence of secreted glycoprotein Chitinase-3-like 1 (CHI3L1) was shown to increase pneumococcal replication in mice. We therefore hypothesized that an IAV preinfection decreases CHI3L1 levels to promote pneumococcal infection. Indeed, in an air-liquid interface model of primary human bronchial epithelial cells (hBECs), IAV preinfection interfered with apical but not basolateral CHI3L1 release. Confocal time-lapse microscopy revealed that the gradual loss of apical CHI3L1 localization during co-infection with influenza and S. pneumoniae coincided with the disappearance of goblet as well as ciliated cells and increased S. pneumoniae replication. Importantly, extracellular restoration of CHI3L1 levels using recombinant protein significantly reduced bacterial load in influenza preinfected bronchial models. Thus, recombinant CHI3L1 may provide a novel therapeutic means to lower morbidity and mortality associated with post-influenza pneumococcal infections.
Collapse
Affiliation(s)
- Diana Karwelat
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany
| | - Bernd Schmeck
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany.,Department of Pulmonary and Critical Care Medicine, University Medical Center Marburg, Universities of Giessen and Marburg Lung Center, Philipps University Marburg, Hesse, Germany.,German Center for Lung Research (DZL), Marburg, Hesse, Germany.,Center for Synthetic Microbiology (SYNMIKRO), Philipps University Marburg, Marburg, Hesse, Germany
| | - Marc Ringel
- Institute of Virology, Philipps University Marburg, Marburg, Hesse, Germany
| | - Birke J Benedikter
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany
| | - Kathleen Hübner
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany
| | - Isabell Beinborn
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany
| | - Andrea Maisner
- Institute of Virology, Philipps University Marburg, Marburg, Hesse, Germany
| | - Leon N Schulte
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany.,German Center for Lung Research (DZL), Marburg, Hesse, Germany
| | - Evelyn Vollmeister
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany
| |
Collapse
|
36
|
Hussain MT, Iqbal AJ, Norling LV. The Role and Impact of Extracellular Vesicles in the Modulation and Delivery of Cytokines during Autoimmunity. Int J Mol Sci 2020; 21:E7096. [PMID: 32993051 PMCID: PMC7584003 DOI: 10.3390/ijms21197096] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
Cytokines and extracellular vesicles are two methods of initiating and maintaining cellular crosstalk. The role of cytokines in the initiation, progression, and resolution of inflammation has been well studied and more so, their pathophysiological role in the development of autoimmune disease. In recent years, the impact of extracellular vesicles on the progression of autoimmunity has become more widely appreciated. In this review, we discuss the mechanisms that allow extracellular vesicles of various sources to modulate cytokine production, and release, and how extracellular vesicles might be involved in the direct delivery and modulation of cytokine levels. Moreover, we explore what challenges are faced by current therapies and the promising future for extracellular vesicles as therapeutic agents in conditions driven by immune dysregulation.
Collapse
Affiliation(s)
- Mohammed Tayab Hussain
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London E1 4NS, UK;
| | - Asif Jilani Iqbal
- The Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Lucy Victoria Norling
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London E1 4NS, UK;
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London E1 4NS, UK
| |
Collapse
|
37
|
Ahmad JN, Sebo P. Adenylate Cyclase Toxin Tinkering With Monocyte-Macrophage Differentiation. Front Immunol 2020; 11:2181. [PMID: 33013916 PMCID: PMC7516048 DOI: 10.3389/fimmu.2020.02181] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/10/2020] [Indexed: 01/28/2023] Open
Abstract
Circulating inflammatory monocytes are attracted to infected mucosa and differentiate into macrophage or dendritic cells endowed with enhanced bactericidal and antigen presenting capacities. In this brief Perspective we discuss the newly emerging insight into how the cAMP signaling capacity of Bordetella pertussis adenylate cyclase toxin manipulates the differentiation of monocytes and trigger dedifferentiation of the alveolar macrophages to facilitate bacterial colonization of human airways.
Collapse
Affiliation(s)
- Jawid Nazir Ahmad
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the CAS, Prague, Czechia
| | - Peter Sebo
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the CAS, Prague, Czechia
| |
Collapse
|
38
|
Barnes BJ, Somerville CC. Modulating Cytokine Production via Select Packaging and Secretion From Extracellular Vesicles. Front Immunol 2020; 11:1040. [PMID: 32547552 PMCID: PMC7272603 DOI: 10.3389/fimmu.2020.01040] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
Cytokines are soluble factors that play vital roles in systemic function due to their ability to initiate and mediate cell-to-cell communication. Another important mechanism of intercellular communication that has gained significant attention in the past 10 years is the release of extracellular vesicles (EVs). EVs are released by all cells during normal physiology, in states of resting and activation, as well as during disease. Accumulating evidence indicates that cytokines may be packaged into EVs, and the packaging of cytokines into EVs, along with their ultimate secretion, may also be regulated by cytokines. Importantly, the repertoire of biomolecules packaged into EVs is shaped by the biological state of the cell (resting vs. activated and healthy vs. disease) and the EV biogenesis pathway involved, thus providing mechanisms by which EV packaging and secretion may be modulated. Given the critical role of cytokines in driving acute and chronic inflammatory and autoimmune diseases, as well as their role in establishing the tumor immune microenvironment, in this review, we will focus on these disease settings and summarize recent progress and mechanisms by which cytokines may be packaged within and modulated by EVs, as a therapeutic option for regulating innate and adaptive immunity.
Collapse
Affiliation(s)
- Betsy J Barnes
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, United States.,Departments of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, United States
| | - Carter C Somerville
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| |
Collapse
|
39
|
In Vitro Effects of Streptococcus oralis Biofilm on Peri-Implant Soft Tissue Cells. Cells 2020; 9:cells9051226. [PMID: 32429151 PMCID: PMC7290395 DOI: 10.3390/cells9051226] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Human gingival epithelial cells (HGEps) and fibroblasts (HGFs) are the main cell types in peri-implant soft tissue. HGEps are constantly exposed to bacteria, but HGFs are protected by connective tissue as long as the mucosa-implant seal is intact. Streptococcus oralis is one of the commensal bacteria, is highly abundant at healthy implant sites, and might modulate soft tissue cells-as has been described for other streptococci. We have therefore investigated the effects of the S. oralis biofilm on HGEps and HGFs. HGEps or HGFs were grown separately on titanium disks and responded to challenge with S. oralis biofilm. HGFs were severely damaged after 4 h, exhibiting transcriptional inflammatory and stress responses. In contrast, challenge with S. oralis only induced a mild transcriptional inflammatory response in HGEps, without cellular damage. HGFs were more susceptible to the S. oralis biofilm than HGEps. The pro-inflammatory interleukin 6 (IL-6) was attenuated in HGFs, as was interleukin 8 (CXCL8) in HGEps. This indicates that S. oralis can actively protect tissue. In conclusion, commensal biofilms can promote homeostatic tissue protection, but only if the implant-mucosa interface is intact and HGFs are not directly exposed.
Collapse
|
40
|
Duncan SE, Gao S, Sarhene M, Coffie JW, Linhua D, Bao X, Jing Z, Li S, Guo R, Su J, Fan G. Macrophage Activities in Myocardial Infarction and Heart Failure. Cardiol Res Pract 2020; 2020:4375127. [PMID: 32377427 PMCID: PMC7193281 DOI: 10.1155/2020/4375127] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/21/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023] Open
Abstract
Heart diseases remain the major cause of death worldwide. Advances in pharmacological and biomedical management have resulted in an increasing proportion of patients surviving acute heart failure (HF). However, many survivors of HF in the early stages end up increasing the disease to chronic HF (CHF). HF is an established frequent complication of myocardial infarction (MI), and numerous influences including persistent myocardial ischemia, shocked myocardium, ventricular remodeling, infarct size, and mechanical impairments, as well as hibernating myocardium trigger the development of left ventricular systolic dysfunction following MI. Macrophage population is active in inflammatory process, yet the clear understanding of the causative roles for these macrophage cells in HF development and progression is actually incomplete. Long ago, it was thought that macrophages are of importance in the heart after MI. Also, though inflammation is as a result of adverse HF in patients, but despite the fact that broad immunosuppression therapeutic target has been used in various clinical trials, no positive results have showed up, but rather, the focus on proinflammatory cytokines has proved more benefits in patients with HF. Therefore, in this review, we discuss the recent findings and new development about macrophage activations in HF, its role in the healthy heart, and some therapeutic targets for myocardial repair. We have a strong believe that there is a need to give maximum attention to cardiac resident macrophages due to the fact that they perform various tasks in wound healing, self-renewal of the heart, and tissue remodeling. Currently, it has been discovered that the study of macrophages goes far beyond its phagocytotic roles. If researchers in future confirm that macrophages play a vital role in the heart, they can be therapeutically targeted for cardiac healing.
Collapse
Affiliation(s)
- Sophia Esi Duncan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Shan Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Michael Sarhene
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Joel Wake Coffie
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Deng Linhua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Xingru Bao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Zhang Jing
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Sheng Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Rui Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Jing Su
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| |
Collapse
|
41
|
Hao Y, Dong X, Zhang M, Liu H, Zhu L, Wang Y. Effects of hyperbaric oxygen therapy on the expression levels of the inflammatory factors interleukin-12p40, macrophage inflammatory protein-1β, platelet-derived growth factor-BB, and interleukin-1 receptor antagonist in keloids. Medicine (Baltimore) 2020; 99:e19857. [PMID: 32312010 PMCID: PMC7220187 DOI: 10.1097/md.0000000000019857] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Our study aimed to screen and explore the expression of inflammatory factors in keloid patients and to investigate how hyperbaric oxygen (HBO) therapy affects the expression levels of interleukin-12p40 (IL-12p40), macrophage inflammatory protein-1β (MIP-1β), platelet-derived growth factor-BB (PDGF-BB), and interleukin-1 receptor antagonist (IL-1Ra). OBJECTIVE 30 patients were randomly selected and divided into the following 3 groups: keloid samples from keloid patients treated with HBO therapy (A), keloid samples from keloid patients treated without HBO therapy (B), and normal control skin samples derived from individuals who had no clear scarring (C). Each group included 10 samples. METHODS Inflammatory factors in the keloid tissues were measured with the MILLIPLEX multiplexed Luminex system. Hematoxylin and eosin staining, immunohistochemical staining, and Western blotting were used to observe the morphological differences in different tissues and the expression levels. RESULTS The expression levels of inflammatory mediators, including IL-12p40, MIP-1β, PDGF-BB, and IL-1Ra, in keloid tissues were significantly different from those in samples of normal skin. Hematoxylin and eosin staining showed significantly greater inflammatory infiltration in keloid tissue. Significantly different expression levels were observed in group A, B, and C. CONCLUSION Significantly altered levels of inflammatory factors in the samples from keloid patients were observed, suggesting that formation of a keloid is potentially related to inflammatory responses. HBO therapy could significantly affect the expression levels of IL-12p40, MIP-1β, PDGF-BB, and IL-1Ra, indicating that the effects of HBO therapy are associated with the attenuation of inflammatory responses.
Collapse
Affiliation(s)
- Yan Hao
- Department of Plastic Surgery, Peking Union Medical College Hospital
| | - Xinhang Dong
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Mingzi Zhang
- Department of Plastic Surgery, Peking Union Medical College Hospital
| | - Hao Liu
- Department of Plastic Surgery, Peking Union Medical College Hospital
| | - Lin Zhu
- Department of Plastic Surgery, Peking Union Medical College Hospital
| | - Youbin Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital
| |
Collapse
|
42
|
Autophagy and Macrophage Functions: Inflammatory Response and Phagocytosis. Cells 2019; 9:cells9010070. [PMID: 31892110 PMCID: PMC7016593 DOI: 10.3390/cells9010070] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/15/2019] [Accepted: 12/20/2019] [Indexed: 12/24/2022] Open
Abstract
Autophagy is a conserved bulk degradation and recycling process that plays important roles in multiple biological functions, including inflammatory responses. As an important component of the innate immune system, macrophages are involved in defending cells from invading pathogens, clearing cellular debris, and regulating inflammatory responses. During the past two decades, accumulated evidence has revealed the intrinsic connection between autophagy and macrophage function. This review focuses on the role of autophagy, both as nonselective and selective forms, in the regulation of the inflammatory and phagocytotic functions of macrophages. Specifically, the roles of autophagy in pattern recognition, cytokine release, inflammasome activation, macrophage polarization, LC3-associated phagocytosis, and xenophagy are comprehensively reviewed. The roles of autophagy receptors in the macrophage function regulation are also summarized. Finally, the obstacles and remaining questions regarding the molecular regulation mechanisms, disease association, and therapeutic applications are discussed.
Collapse
|
43
|
Aiello A, Giannessi F, Percario ZA, Affabris E. An emerging interplay between extracellular vesicles and cytokines. Cytokine Growth Factor Rev 2019; 51:49-60. [PMID: 31874738 DOI: 10.1016/j.cytogfr.2019.12.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/17/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs) are small membrane-bound particles that are naturally released from cells. They are recognized as potent vehicles of intercellular communication both in prokaryotes and eukaryotes. Because of their capacity to carry biological macromolecules such as proteins, lipids and nucleic acids, EVs influence different physiological and pathological functions of both parental and recipient cells. Although multiple pathways have been proposed for cytokine secretion beyond the classical ER/Golgi route, EVs have recently recognized as an alternative secretory mechanism. Interestingly, cytokines/chemokines exploit these vesicles to be released into the extracellular milieu, and also appear to modulate their release, trafficking and/or content. In this review, we provide an overview of the cytokines/chemokines that are known to be associated with EVs or their regulation with a focus on TNFα, IL-1β and IFNs.
Collapse
|
44
|
Giovos G, Yavropoulou MP, Yovos JG. The role of cellular senescence in diabetes mellitus and osteoporosis: molecular pathways and potential interventions. Hormones (Athens) 2019; 18:339-351. [PMID: 31701490 DOI: 10.1007/s42000-019-00132-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023]
Abstract
The improving effectiveness of health care leads inevitably to a rapid increase in the elderly population worldwide. At advanced ages, however, people experience chronic disabilities, which significantly increase the social and economic burden while curtailing survival, independence, and quality of life of the aging population. As aging is a multifactorial process, apart from genetic predisposition, other environmental factors, such as chronic sterile inflammation and cellular senescence, contribute as crucial participants and have been targeted to reverse their deleterious effects on tissue homeostasis and functional integrity. Cellular senescence refers to the essentially irreversible inhibition of cellular proliferation when cells are subjected to extrinsic or endogenous stress. Although the process of cellular senescence has long been known, recent evidence demonstrated that it characterizes many aging phenotypes and that elimination of senescent cells at the tissue level can improve age-related tissue dysfunction. These observations have renewed scientific interest in possible therapeutic interventions. Two major chronic diseases associated with aging that impose an enormous burden on global health systems are type 2 diabetes and osteoporosis. This review presents current data on (i) the underlying molecular mechanisms of cellular senescence, (ii) its relationship to these two endocrine diseases that are today prevalent worldwide, and (iii) future prospects of targeted intervention with the aim of simultaneously improving the progression and prognosis of these serious problems of aging.
Collapse
Affiliation(s)
- Georgios Giovos
- Clinical Research Fellow in Endocrinology, Wisdem Centre, University Hospitals Coventry & Warwickshire, Coventry, UK
| | - Maria P Yavropoulou
- Endocrinology Unit, 1st Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, UOA, LAIKO General Hospital, 17 Agiou Thoma Str., 11527, Athens, Greece.
| | - John G Yovos
- Professor Emeritus in Internal Medicine and Endocrinology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
45
|
Sprooten J, Garg AD. Type I interferons and endoplasmic reticulum stress in health and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 350:63-118. [PMID: 32138904 PMCID: PMC7104985 DOI: 10.1016/bs.ircmb.2019.10.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type I interferons (IFNs) comprise of pro-inflammatory cytokines created, as well as sensed, by all nucleated cells with the main objective of blocking pathogens-driven infections. Owing to this broad range of influence, type I IFNs also exhibit critical functions in many sterile inflammatory diseases and immunopathologies, especially those associated with endoplasmic reticulum (ER) stress-driven signaling pathways. Indeed, over the years accumulating evidence has indicated that the presence of ER stress can influence the production, or sensing of, type I IFNs induced by perturbations like pattern recognition receptor (PRR) agonists, infections (bacterial, viral or parasitic) or autoimmunity. In this article we discuss the link between type I IFNs and ER stress in various diseased contexts. We describe how ER stress regulates type I IFNs production or sensing, or how type I IFNs may induce ER stress, in various circumstances like microbial infections, autoimmunity, diabetes, cancer and other ER stress-related contexts.
Collapse
Affiliation(s)
- Jenny Sprooten
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium.
| |
Collapse
|
46
|
Cho DI, Kang HJ, Jeon JH, Eom GH, Cho HH, Kim MR, Cho M, Jeong HY, Cho HC, Hong MH, Kim YS, Ahn Y. Antiinflammatory activity of ANGPTL4 facilitates macrophage polarization to induce cardiac repair. JCI Insight 2019; 4:125437. [PMID: 31434807 DOI: 10.1172/jci.insight.125437] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 07/23/2019] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can suppress pathological inflammation. However, the mechanisms underlying the association between MSCs and inflammation remain unclear. Under coculture conditions with macrophages, MSCs highly expressed angiopoietin-like 4 (ANGPTL4) to blunt the polarization of macrophages toward the proinflammatory phenotype. ANGPTL4-deficient MSCs failed to inhibit the inflammatory macrophage phenotype. In inflammation-related animal models, the injection of coculture medium or ANGPTL4 protein increased the antiinflammatory macrophages in both peritonitis and myocardial infarction. In particular, cardiac function and pathology were markedly improved by ANGPTL4 treatment. We found that retinoic acid-related orphan receptor α (RORα) was increased by inflammatory mediators, such as IL-1β, and bound to ANGPTL4 promoter in MSCs. Collectively, RORα-mediated ANGPTL4 induction was shown to contribute to the antiinflammatory activity of MSCs against macrophages under pathological conditions. This study suggests that the capability of ANGPTL4 to induce tissue repair is a promising opportunity for safe stem cell-free regeneration therapy from a translational perspective.
Collapse
Affiliation(s)
- Dong Im Cho
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Hye-Jin Kang
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Ju Hee Jeon
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Gwang Hyeon Eom
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Korea
| | - Hyang Hee Cho
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea.,Department of Molecular Medicine, Graduate School, Chonnam National University, Gwangju, Korea
| | - Mi Ra Kim
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Meeyoung Cho
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Hye-Yun Jeong
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Hyen Chung Cho
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea.,Department of Molecular Medicine, Graduate School, Chonnam National University, Gwangju, Korea
| | - Moon Hwa Hong
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Yong Sook Kim
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea.,Biomedical Research Institute, Chonnam National University Hospital, Gwangju, Korea
| | - Youngkeun Ahn
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea.,Department of Cardiology, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
47
|
Loktionov A. Eosinophils in the gastrointestinal tract and their role in the pathogenesis of major colorectal disorders. World J Gastroenterol 2019; 25:3503-3526. [PMID: 31367153 PMCID: PMC6658389 DOI: 10.3748/wjg.v25.i27.3503] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/22/2019] [Accepted: 06/01/2019] [Indexed: 02/06/2023] Open
Abstract
Eosinophils are currently regarded as versatile mobile cells controlling and regulating multiple biological pathways and responses in health and disease. These cells store in their specific granules numerous biologically active substances (cytotoxic cationic proteins, cytokines, growth factors, chemokines, enzymes) ready for rapid release. The human gut is the main destination of eosinophils that are produced and matured in the bone marrow and then transferred to target tissues through the circulation. In health the most important functions of gut-residing eosinophils comprise their participation in the maintenance of the protective mucosal barrier and interactions with other immune cells in providing immunity to microbiota of the gut lumen. Eosinophils are closely involved in the development of inflammatory bowel disease (IBD), when their cytotoxic granule proteins cause damage to host tissues. However, their roles in Crohn’s disease and ulcerative colitis appear to follow different immune response patterns. Eosinophils in IBD are especially important in altering the structure and protective functions of the mucosal barrier and modulating massive neutrophil influx to the lamina propria followed by transepithelial migration to colorectal mucus. IBD-associated inflammatory process involving eosinophils then appears to expand to the mucus overlaying the internal gut surface. The author hypothesises that immune responses within colorectal mucus as well as ETosis exerted by both neutrophils and eosinophils on the both sides of the colonic epithelial barrier act as additional pathogenetic factors in IBD. Literature analysis also shows an association between elevated eosinophil levels and better colorectal cancer (CRC) prognosis, but mechanisms behind this effect remain to be elucidated. In conclusion, the author emphasises the importance of investigating colorectal mucus in IBD and CRC patients as a previously unexplored milieu of disease-related inflammatory responses.
Collapse
|
48
|
Role of microglial activation and neuroinflammation in neurotoxicity of acrylamide in vivo and in vitro. Arch Toxicol 2019; 93:2007-2019. [PMID: 31073625 DOI: 10.1007/s00204-019-02471-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
Acrylamide, a soft electrophile, is widely used in the industry and laboratories, and also contaminates certain foods. Neurotoxicity and neurodegenerative effects of acrylamide have been reported in humans and experimental animals, although the underlying mechanism remains obscure. Activation of microglia and neuroinflammation has been demonstrated in various neurodegenerative diseases as well as other pathologies of the brain. The present study aimed to investigate the role of microglial activation and neuroinflammation in acrylamide neurotoxicity. Male 10-week-old Wistar rats were exposed to acrylamide by gavage at 0, 0.2, 2, or 20 mg/kg BW, once per day for 5 weeks. The results showed that 5-week exposure to acrylamide induced inflammatory responses in the cerebral cortex, evident by upregulated mRNA and protein expression of pro-inflammatory cytokines IL-1β, IL-6, and IL-18. Acrylamide also induced activation of microglia, indicated by increased expression of microglial markers, CD11b and CD40, and increased CD11b/c-positive microglial area and microglial process length. In vitro studies using BV-2 microglial cells confirmed microglial inflammatory response, as evident by time- (0-36 h; 50 μM) and dose- (0-500 μM; 24 h) dependent increase in mRNA expression of IL-1β and IL-18, as well as the inflammatory marker iNOS. Furthermore, acrylamide-induced upregulation of pro-inflammatory cytokines was mediated through the NLRP3 inflammasome pathway, as evident by increased expression of NLRP3, caspase 1, and ASC in the rat cerebral cortex, and by the inhibitory effects of NLRP3 inflammasome inhibitor on the acrylamide-induced upregulation of NLRP3, caspase 1, IL-1β, and IL-18 in BV-2 microglia.
Collapse
|
49
|
Infection by the parasitic helminth Trichinella spiralis activates a Tas2r-mediated signaling pathway in intestinal tuft cells. Proc Natl Acad Sci U S A 2019; 116:5564-5569. [PMID: 30819885 PMCID: PMC6431192 DOI: 10.1073/pnas.1812901116] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Intestinal tuft cells are sentinels monitoring the luminal contents and play a critical role in type 2 immunity. In this work, Trichinella spiralis excretion–secretion and extract were shown to directly induce interleukin 25 (IL-25) release from the intestinal villi, evoke calcium responses in tuft cells, and activate Tas2r bitter-taste receptors, whereas the bitter compound salicin was shown to activate and induce tuft cells to release IL-25. Gα-gustducin/Gβ1γ13 and/or Gαo/Gβ1γ13, Plcβ2, Ip3r2, and Trpm5 comprise the signal transduction pathways that tuft cells utilize to initiate type 2 immune responses. Potentiation of Trpm5 by a natural sweet compound, stevioside, can enhance the tuft cell–ILC2 circuit’s activity, indicating that modulating these signaling components can help devise new means of combating parasites. The parasitic helminth Trichinella spiralis, which poses a serious health risk to animals and humans, can be found worldwide. Recent findings indicate that a rare type of gut epithelial cell, tuft cells, can detect the helminth, triggering type 2 immune responses. However, the underlying molecular mechanisms remain to be fully understood. Here we show that both excretory–secretory products (E–S) and extract of T. spiralis can stimulate the release of the cytokine interleukin 25 (IL-25) from the mouse small intestinal villi and evoke calcium responses from tuft cells in the intestinal organoids, which can be blocked by a bitter-taste receptor inhibitor, allyl isothiocyanate. Heterologously expressed mouse Tas2r bitter-taste receptors, the expression of which is augmented during tuft-cell hyperplasia, can respond to the E–S and extract as well as to the bitter compound salicin whereas salicin in turn can induce IL-25 release from tuft cells. Furthermore, abolishment of the G-protein γ13 subunit, application of the inhibitors for G-protein αo/i, Gβγ subunits, and phospholipase Cβ2 dramatically reduces the IL-25 release. Finally, tuft cells are found to utilize the inositol triphosphate receptor type 2 (Ip3r2) to regulate cytosolic calcium and thus Trpm5 activity, while potentiation of Trpm5 by a sweet-tasting compound, stevioside, enhances tuft cell IL-25 release and hyperplasia in vivo. Taken together, T. spiralis infection activates a signaling pathway in intestinal tuft cells similar to that of taste-bud cells, but with some key differences, to initiate type 2 immunity.
Collapse
|
50
|
VAMP8, a vesicle-SNARE required for RAB37-mediated exocytosis, possesses a tumor metastasis suppressor function. Cancer Lett 2018; 437:79-88. [PMID: 30165196 DOI: 10.1016/j.canlet.2018.08.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 08/20/2018] [Indexed: 12/23/2022]
Abstract
We previously identified a metastasis suppressor RAB37 small GTPase that regulated exocytosis of tissue inhibitor of metalloproteinases 1 (TIMP1) to suppress lung cancer metastasis. Here, we show that vesicle-associated membrane protein 8 (VAMP8), a v-SNARE (vesicle soluble N-ethylmaleimide-sensitive factor activating protein receptor), interacts with RAB37 and drives the secretion of TIMP1 to inhibit tumor metastases. Confocal and total internal reflection fluorescence microscopic images demonstrated that VAMP8 co-localized with RAB37 and facilitated trafficking of RAB37-TIMP1 vesicles. Reconstitution experiments using tail-vein injection and lung-to-lung metastasis in mice showed that VAMP8 was essential for RAB37-regulated vesicle trafficking of TIMP1 to suppress cancer metastasis. Lung cancer patients with low VAMP8 showed distant metastasis, poor overall survival and progression-free survival. Importantly, multivariate Cox regression analysis indicated that patients with low VAMP8/low RAB37 expression profile showed significantly high risk of death (hazard ratio = 3.42, P < 0.001) even after adjusting for tumor metastasis parameter. Our findings reveal that VAMP8 is a novel v-SNARE crucial for RAB37-mediated exocytic transport of TIMP1 to suppress lung tumor metastasis. VAMP8 possesses a tumor metastasis suppressor function with a prognostic value in lung cancer.
Collapse
|