1
|
Lee SY, Lee BY, Lim B, Uzzaman R, Jang G, Kim KS. Exploring the importance of predicted camel NRAP exon 4 for environmental adaptation using a mouse model. Anim Genet 2025; 56:e13490. [PMID: 39478283 DOI: 10.1111/age.13490] [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: 04/26/2024] [Revised: 09/28/2024] [Accepted: 10/20/2024] [Indexed: 12/25/2024]
Abstract
Camels possess exceptional adaptability, allowing them to withstand extreme temperatures in desert environments. They conserve water by reducing their metabolic rate and regulating body temperature. The heart of the camel plays a crucial role in this adaptation, with specific genes expressed in cardiac tissue that are essential for mammalian adaptation, regulating cardiac function and responding to environmental stressors. One such gene, nebulin-related-anchoring protein (NRAP), is involved in the assembly of myofibrils and the transmission of force within the heart. In our study of the NRAP gene across various livestock species, including three camel species, we identified a camel-specific exon region in the NRAP transcripts. This additional exon (exon 4) contains an open reading frame predicted in camels. To investigate its function, we generated knock-in mice expressing camel NRAP exon 4. These 'camelized mice' exhibited normal phenotypic characteristics compared with wild-type mice but showed elevated body temperatures under cold stress. Transcriptome analyses of the hearts from camelized mice under cold stress revealed differentially expressed inflammatory cytokine genes, known to influence cardiac function by modulating the contractility of cardiac muscle cells. We propose further investigations utilizing these camelized mice to explore these findings in greater depth.
Collapse
Affiliation(s)
- Sung-Yeon Lee
- Department of Animal Sciences, Chungbuk National University, Cheongju, South Korea
- GEM Division, Macrogen Inc., Seoul, South Korea
- Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Bo-Young Lee
- Department of Animal Sciences, Chungbuk National University, Cheongju, South Korea
- Department of Biological Science, University of New Hampshire, Durham, New Hampshire, USA
| | - Byeonghwi Lim
- Department of Animal Science and Technology, Chung-Ang University, Anseong, South Korea
| | - Rasel Uzzaman
- Department of Animal Sciences, Chungbuk National University, Cheongju, South Korea
| | - Goo Jang
- Laboratory of Theriogenology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Kwan-Suk Kim
- Department of Animal Sciences, Chungbuk National University, Cheongju, South Korea
| |
Collapse
|
2
|
Bai L, Gao Y, Li L, Liang Z, Qiao Y, Wang X, Yv L, Yang JJ, Xu JT. Poly-(ADP-ribose) polymerase 1-modulated production of CXCL1 in the dorsal root ganglion and spinal dorsal horn exacerbated inflammatory pain in rats. Int Immunopharmacol 2024; 143:113370. [PMID: 39405941 DOI: 10.1016/j.intimp.2024.113370] [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: 07/09/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024]
Abstract
Poly (ADP-ribose) polymerase 1 (PARP-1) serves as a transcriptional co-regulator and has been playing an important role in various inflammatory diseases. In the present study, we investigated the role and underlying mechanisms of action of PARP-1 in inflammatory pain. Intraplantar injection of complete Freund's adjuvant (CFA) was administered to the rats to induce inflammatory pain. Immunofluorescence, Western blotting, co-immunoprecipitation, and chromatin immunoprecipitation-quantitative polymerase chain reaction were performed to investigate the underlying mechanisms. Our results showed that CFA injection led to an increase in the production and activation of PARP-1 in both the L4/5 dorsal root ganglions (DRGs) and the spinal dorsal horn. Repeated intrathecal injections of Tiq-A or 5-AIQ, two specific inhibitors of PARP-1, and microinjections of AAV-PARP-1 shRNA into the L5 DRG or L5 spinal dorsal horn partially prevented the development of inflammatory pain. The established inflammatory pain was attenuated by a single bolus of intrathecal injection of Tiq-A or 5-AIQ on day 7 after the CFA injection. The CFA-induced mechanical allodynia and thermal hyperalgesia in female rats were alleviated by repeated intrathecal injections of Tiq-A. Moreover, repeated intrathecal injections of 5-AIQ inhibited the binding of NF-κB with CXCL1 promoter and reduced the production of CXCL1 in both the L4/5 DRGs and L4-6 spinal dorsal horns following CFA injection. Collectively, our results indicate that CFA-induced upregulation of PARP-1 by promoting CXCL1 expression in the DRG and probably in the spinal dorsal horn contributes to the pathogenesis of inflammatory pain. Thus, PARP-1 may be a potential pharmaceutical target for the treatment of inflammatory pain.
Collapse
Affiliation(s)
- Liying Bai
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Yan Gao
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China; Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Zongyi Liang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Yiming Qiao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Xueli Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Lili Yv
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China; Neuroscience Research Institute, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China.
| | - Ji-Tian Xu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China; Neuroscience Research Institute, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China.
| |
Collapse
|
3
|
Torelli FR, Rodrigues-Peres RM, Monteiro I, Lopes-Cendes I, Bahamondes L, Juliato CRT. Gene expression associated with unfavorable vaginal bleeding in women using the etonogestrel subdermal contraceptive implant: a prospective study. Sci Rep 2024; 14:11062. [PMID: 38745005 PMCID: PMC11093992 DOI: 10.1038/s41598-024-61751-7] [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: 12/13/2023] [Accepted: 05/09/2024] [Indexed: 05/16/2024] Open
Abstract
To evaluate gene expression associated with unfavorable vaginal bleeding in users of the Etonogestrel (ENG) contraceptive implant. Prospective study involving 100 women who intended to use the ENG implant. Exclusion criteria included abnormal uterine bleeding, inability to attend a 1-year follow-up, and implant removal for reasons unrelated to vaginal bleeding or loss of follow-up. We obtained endometrial biopsies before implant placement and assessed the expression of 20 selected genes. Users maintained a uterine bleeding diary for 12 months post-implant placement. For statistical analysis, we categorized women into those with or without favorable vaginal bleeding at 3 and 12 months. Women with lower CXCL1 expression had a 6.8-fold increased risk of unfavorable vaginal bleeding at 3 months (OR 6.8, 95% CI 2.21-20.79, p < 0.001), while those with higher BCL6 and BMP6 expression had 6- and 5.1-fold increased risks, respectively. By the 12-month follow-up, women with lower CXCL1 expression had a 5.37-fold increased risk of unfavorable vaginal bleeding (OR 5.37, 95% CI 1.63-17.73, p = 0.006). Women with CXCL1 expression < 0.0675, BCL6 > 0.65, and BMP6 > 3.4 had a higher likelihood of experiencing unfavorable vaginal bleeding at 3 months, and CXCL1 < 0.158 at 12 months. Users of ENG contraceptive implants with elevated BCL6 and BMP6 expression exhibited a higher risk of breakthrough bleeding at the 3-month follow-up. Conversely, reduced CXCL1 expression was associated with an elevated risk of bleeding at both the 3 and 12-month follow-ups.
Collapse
Affiliation(s)
- Flávia R Torelli
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Raquel M Rodrigues-Peres
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Ilza Monteiro
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Iscia Lopes-Cendes
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Luis Bahamondes
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Cássia R T Juliato
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil.
- Departamento de Tocoginecologia, Faculdade de Ciências Médicas, Universidade Estadual de Campinas - UNICAMP, Rua Alexander Fleming 101, Campinas, SP, 13083-881, Brazil.
| |
Collapse
|
4
|
Zhang L, Cascio S, Mellors JW, Buckanovich RJ, Osmanbeyoglu HU. Single-cell analysis reveals the stromal dynamics and tumor-specific characteristics in the microenvironment of ovarian cancer. Commun Biol 2024; 7:20. [PMID: 38182756 PMCID: PMC10770164 DOI: 10.1038/s42003-023-05733-x] [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: 06/22/2023] [Accepted: 12/20/2023] [Indexed: 01/07/2024] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is a heterogeneous disease, and a highstromal/desmoplastic tumor microenvironment (TME) is associated with a poor outcome. Stromal cell subtypes, including fibroblasts, myofibroblasts, and cancer-associated mesenchymal stem cells, establish a complex network of paracrine signaling pathways with tumor-infiltrating immune cells that drive effector cell tumor immune exclusion and inhibit the antitumor immune response. In this work, we integrate single-cell transcriptomics of the HGSOC TME from public and in-house datasets (n = 20) and stratify tumors based upon high vs. low stromal cell content. Although our cohort size is small, our analyses suggest a distinct transcriptomic landscape for immune and non-immune cells in high-stromal vs. low-stromal tumors. High-stromal tumors have a lower fraction of certain T cells, natural killer (NK) cells, and macrophages, and increased expression of CXCL12 in epithelial cancer cells and cancer-associated mesenchymal stem cells (CA-MSCs). Analysis of cell-cell communication indicate that epithelial cancer cells and CA-MSCs secrete CXCL12 that interacte with the CXCR4 receptor, which is overexpressed on NK and CD8+ T cells. Dual IHC staining show that tumor infiltrating CD8 T cells localize in proximity of CXCL12+ tumor area. Moreover, CXCL12 and/or CXCR4 antibodies confirm the immunosuppressive role of CXCL12-CXCR4 in high-stromal tumors.
Collapse
Affiliation(s)
- Linan Zhang
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15206, USA
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA
- Department of Applied Mathematics, School of Mathematics and Statistics, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Sandra Cascio
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA
- Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - John W Mellors
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Ronald J Buckanovich
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA
- Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15232, USA
| | - Hatice Ulku Osmanbeyoglu
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15206, USA.
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.
- Department of Bioengineering, University of Pittsburgh School of Engineering, Pittsburgh, PA, 15219, USA.
- Department of Biostatistics, University of Pittsburgh School of Public Health, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
5
|
Qiu XN, Hong D, Shi ZR, Lu SY, Lai YX, Ren YL, Liu XT, Guo CP, Tan GZ, Wang LC. TNF-α promotes CXCL-1/8 production in keratinocytes by downregulating galectin-3 through NF-κB and hsa-miR-27a-3p pathway to contribute psoriasis development. Immunopharmacol Immunotoxicol 2023; 45:692-700. [PMID: 37358143 DOI: 10.1080/08923973.2023.2229510] [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: 07/23/2022] [Accepted: 06/18/2023] [Indexed: 06/27/2023]
Abstract
OBJECTIVE Treatment with TNF-α inhibitors improve psoriasis with minimize/minor neutrophils infiltration and CXCL-1/8 expression in psoriatic lesions. However, the fine mechanism of TNF-α initiating psoriatic inflammation by tuning keratinocytes is unclear. Our previous research identified the deficiency of intracellular galectin-3 was sufficient to promote psoriasis inflammation characterized by neutrophil accumulation. This study aims to investigate whether TNF-α participated in psoriasis development through dysregulating galectin-3 expression. METHODS mRNA levels were assessed through quantitative real-time PCR. Flow cytometry was used to detect cell cycle/apoptosis. Western blot was used to evaluate the activation of the NF-κB signaling pathway. HE staining and immunochemistry were used to detect epidermal thickness and MPO expression, respectively. Specific small interfering RNA (siRNA) was used to knock down hsa-miR-27a-3p while plasmids transfection was used to overexpress galectin-3. Further, the multiMiR R package was utilized to predict microRNA-target interaction. RESULTS AND DISCUSSION We found that TNF-α stimulation altered cell proliferation and differentiation and promoted the production of psoriasis-related inflammatory mediators along with the inhibition of galectin-3 expression in keratinocytes. Supplement of galectin-3 could counteract the rise of CXCL-1/8 but not the other phenotypes of keratinocytes induced by TNF-α. Mechanistically, inhibition of the NF-κB signaling pathway could counteract the decrease of galectin-3 and the increase of hsa-miR-27a-3p expression whereas silence of hsa-miR-27a-3p could counteract the decrease of galectin-3 expression induced by TNF-α treatment in keratinocytes. Intradermal injection of murine anti-CXCL-2 antibody greatly alleviated imiquimod-induced psoriasis-like dermatitis. CONCLUSION TNF-α initiates psoriatic inflammation by increasing CXCL-1/8 in keratinocytes mediated by the axis of NF-κB-hsa-miR-27a-3p-galectin-3 pathway.
Collapse
Affiliation(s)
- Xiao-Nan Qiu
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dan Hong
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhen-Rui Shi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Si-Yao Lu
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu-Xian Lai
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan-Ling Ren
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiu-Ting Liu
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chi-Peng Guo
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guo-Zhen Tan
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liang-Chun Wang
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
6
|
Zhang L, Cascio S, Mellors JW, Buckanovich RJ, Osmanbeyoglu HU. Single-cell analysis reveals the stromal dynamics and tumor-specific characteristics in the microenvironment of ovarian cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544095. [PMID: 37333262 PMCID: PMC10274812 DOI: 10.1101/2023.06.07.544095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
High-grade serous ovarian carcinoma (HGSOC) is a heterogeneous disease, and a high stromal/desmoplastic tumor microenvironment (TME) is associated with a poor outcome. Stromal cell subtypes, including fibroblasts, myofibroblasts, and cancer-associated mesenchymal stem cells, establish a complex network of paracrine signaling pathways with tumor-infiltrating immune cells that drive effector cell tumor immune exclusion and inhibit the antitumor immune response. Single-cell transcriptomics of the HGSOC TME from public and in-house datasets revealed a distinct transcriptomic landscape for immune and non-immune cells in high-stromal vs. low-stromal tumors. High-stromal tumors had a lower fraction of certain T cells, natural killer (NK) cells, and macrophages and increased expression of CXCL12 in epithelial cancer cells and cancer-associated mesenchymal stem cells (CA-MSCs). Analysis of cell-cell communication indicated that epithelial cancer cells and CA-MSCs secreted CXCL12 that interacted with the CXCR4 receptor, which was overexpressed on NK and CD8 + T cells. CXCL12 and/or CXCR4 antibodies confirmed the immunosuppressive role of CXCL12-CXCR4 in high-stromal tumors.
Collapse
|
7
|
Hereditary Tyrosinemia Type 1 Mice under Continuous Nitisinone Treatment Display Remnants of an Uncorrected Liver Disease Phenotype. Genes (Basel) 2023; 14:genes14030693. [PMID: 36980965 PMCID: PMC10047938 DOI: 10.3390/genes14030693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
Hereditary tyrosinemia type 1 (HT1) is a genetic disorder of the tyrosine degradation pathway (TIMD) with unmet therapeutic needs. HT1 patients are unable to fully break down the amino acid tyrosine due to a deficient fumarylacetoacetate hydrolase (FAH) enzyme and, therefore, accumulate toxic tyrosine intermediates. If left untreated, they experience hepatic failure with comorbidities involving the renal and neurological system and the development of hepatocellular carcinoma (HCC). Nitisinone (NTBC), a potent inhibitor of the 4-hydroxyphenylpyruvate dioxygenase (HPD) enzyme, rescues HT1 patients from severe illness and death. However, despite its demonstrated benefits, HT1 patients under continuous NTBC therapy are at risk to develop HCC and adverse reactions in the eye, blood and lymphatic system, the mechanism of which is poorly understood. Moreover, NTBC does not restore the enzymatic defects inflicted by the disease nor does it cure HT1. Here, the changes in molecular pathways associated to the development and progression of HT1-driven liver disease that remains uncorrected under NTBC therapy were investigated using whole transcriptome analyses on the livers of Fah- and Hgd-deficient mice under continuous NTBC therapy and after seven days of NTBC therapy discontinuation. Alkaptonuria (AKU) was used as a tyrosine-inherited metabolic disorder reference disease with non-hepatic manifestations. The differentially expressed genes were enriched in toxicological gene classes related to liver disease, liver damage, liver regeneration and liver cancer, in particular HCC. Most importantly, a set of 25 genes related to liver disease and HCC development was identified that was differentially regulated in HT1 vs. AKU mouse livers under NTBC therapy. Some of those were further modulated upon NTBC therapy discontinuation in HT1 but not in AKU livers. Altogether, our data indicate that NTBC therapy does not completely resolves HT1-driven liver disease and supports the sustained risk to develop HCC over time as different HCC markers, including Moxd1, Saa, Mt, Dbp and Cxcl1, were significantly increased under NTBC.
Collapse
|
8
|
Mohamad Zaki NH, Shiota J, Calder AN, Keeley TM, Allen BL, Nakao K, Samuelson LC, Razumilava N. C-X-C motif chemokine ligand 1 induced by Hedgehog signaling promotes mouse extrahepatic bile duct repair after acute injury. Hepatology 2022; 76:936-950. [PMID: 35388502 PMCID: PMC9790600 DOI: 10.1002/hep.32492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/05/2022] [Accepted: 03/25/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS In extrahepatic bile duct (EHBD) cholangiopathies, including primary sclerosing cholangitis, a reactive cholangiocyte phenotype is associated with inflammation and epithelial hyperproliferation. The signaling pathways involved in EHBD injury response are poorly understood. In this study, we investigated the role of Hedgehog (HH) signaling and its downstream effectors in controlling biliary proliferation and inflammation after EHBD injury. APPROACH AND RESULTS Using mouse bile duct ligation as an acute EHBD injury model, we used inhibitory paradigms to uncover mechanisms promoting the proliferative response. HH signaling was inhibited genetically in Gli1-/- mice or by treating wild-type mice with LDE225. The role of neutrophils was tested using chemical (SB225002) and biological (lymphocyte antigen 6 complex locus G6D [Ly6G] antibodies) inhibitors of neutrophil recruitment. The cellular response was defined through morphometric quantification of proliferating cells and CD45+ and Ly6G+ immune cell populations. Key signaling component expression was measured and localized to specific EHBD cellular compartments by in situ hybridization, reporter strain analysis, and immunohistochemistry. Epithelial cell proliferation peaked 24 h after EHBD injury, preceded stromal cell proliferation, and was associated with neutrophil influx. Indian HH ligand expression in the biliary epithelium rapidly increased after injury. HH-responding cells and neutrophil chemoattractant C-X-C motif chemokine ligand 1 (CXCL1) expression mapped to EHBD stromal cells. Inhibition of HH signaling blocked CXCL1 induction, diminishing neutrophil recruitment and the biliary proliferative response to injury. Directly targeting neutrophils by inhibition of the CXCL1/C-X-C motif chemokine receptor 2/Ly6G signaling axis also decreased biliary proliferation. CONCLUSIONS HH-regulated CXCL1 orchestrates the early inflammatory response and biliary proliferation after EHBD injury through complex cellular crosstalk.
Collapse
Affiliation(s)
| | - Junya Shiota
- Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
- Department of Gastroenterology and HepatologyNagasaki UniversityNagasakiJapan
| | - Ashley N. Calder
- Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Theresa M. Keeley
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Benjamin L. Allen
- Department of Cell and Developmental BiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Kazuhiko Nakao
- Department of Gastroenterology and HepatologyNagasaki UniversityNagasakiJapan
| | - Linda C. Samuelson
- Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | | |
Collapse
|
9
|
An Update on the Chemokine System in the Development of NAFLD. Medicina (B Aires) 2022; 58:medicina58060761. [PMID: 35744024 PMCID: PMC9227560 DOI: 10.3390/medicina58060761] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the world. Sustained hepatic inflammation is a key driver of the transition from simple fatty liver to nonalcoholic steatohepatitis (NASH), the more aggressive form of NAFLD. Hepatic inflammation is orchestrated by chemokines, a family of chemoattractant cytokines that are produced by hepatocytes, Kupffer cells (liver resident macrophages), hepatic stellate cells, endothelial cells, and vascular smooth muscle cells. Over the last three decades, accumulating evidence from both clinical and experimental investigations demonstrated that chemokines and their receptors are increased in the livers of NAFLD patients and that CC chemokine ligand (CCL) 2 and CCL5 in particular play a pivotal role in inducing insulin resistance, steatosis, inflammation, and fibrosis in liver disease. Cenicriviroc (CVC), a dual antagonist of these chemokines’ receptors, CCR2 and CCR5, has been tested in clinical trials in patients with NASH-associated liver fibrosis. Additionally, recent studies revealed that other chemokines, such as CCL3, CCL25, CX3C chemokine ligand 1 (CX3CL1), CXC chemokine ligand 1 (CXCL1), and CXCL16, can also contribute to the pathogenesis of NAFLD. Here, we review recent updates on the roles of chemokines in the development of NAFLD and their blockade as a potential therapeutic approach.
Collapse
|
10
|
Identification of the Resveratrol Potential Targets in the Treatment of Osteoarthritis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9911286. [PMID: 34917160 PMCID: PMC8670923 DOI: 10.1155/2021/9911286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 10/13/2021] [Accepted: 11/20/2021] [Indexed: 11/17/2022]
Abstract
Objectives Osteoarthritis (OA) is a chronic joint degenerative disease and has become an important health problem for the elderly. However, there is still a lack of effective drugs for the treatment of OA. Our research combines bioinformatics and experimental strategies to determine the target of resveratrol for OA treatment. Methods First, the differentially expressed genes (DEGs) of OA joint tissues were obtained from the related microarray gene expression data. Second, resveratrol, a natural polyphenol compound, was used to screen the drug treatment target genes. Third, the drug-disease network was established, and the resveratrol target genes for OA treatment were obtained and verified through experimental verification. Results A total of 300 differentially expressed genes with 246 upregulated and 54 downregulated were found in OA joint tissues, and 310 resveratrol potential target genes were obtained. Finally, six genes, namely, CXCL1, HIF1A, IL-6, MMP3, NOX4, and PTGS2, were selected to validate the treatment effects of the resveratrol. The results showed that all six genes in human OA chondrocytes were significantly increased. In addition, in these chondrocytes, CXCL1, HIF1A, IL-6, MMP3, NOX4, and PTGS2 were reduced considerably, but HIF1A was significantly increased after resveratrol treatment. Conclusions Our data indicates that CXCL1, HIF1A, IL-6, MMP3, NOX4, and PTGS2 are all targets of resveratrol therapy. Our findings may provide valuable information for the mechanism and therapeutic of OA.
Collapse
|
11
|
Alqahtani S, Xia L, Jannasch A, Ferreira C, Franco J, Shannahan JH. Disruption of pulmonary resolution mediators contribute to exacerbated silver nanoparticle-induced acute inflammation in a metabolic syndrome mouse model. Toxicol Appl Pharmacol 2021; 431:115730. [PMID: 34601004 PMCID: PMC8545917 DOI: 10.1016/j.taap.2021.115730] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/08/2021] [Accepted: 09/22/2021] [Indexed: 12/18/2022]
Abstract
Pre-existing conditions modulate sensitivity to numerous xenobiotic exposures such as air pollution. Specifically, individuals suffering from metabolic syndrome (MetS) demonstrate enhanced acute inflammatory responses following particulate matter inhalation. The mechanisms associated with these exacerbated inflammatory responses are unknown, impairing interventional strategies and our understanding of susceptible populations. We hypothesize MetS-associated lipid dysregulation influences mediators of inflammatory resolution signaling contributing to increased acute pulmonary toxicity. To evaluate this hypothesis, healthy and MetS mouse models were treated with either 18-hydroxy eicosapentaenoic acid (18-HEPE), 14-hydroxy docosahexaenoic acid (14-HDHA), 17-hydroxy docosahexaenoic acid (17-HDHA), or saline (control) via intraperitoneal injection prior to oropharyngeal aspiration of silver nanoparticles (AgNP). In mice receiving saline treatment, AgNP exposure resulted in an acute pulmonary inflammatory response that was exacerbated in MetS mice. A targeted lipid assessment demonstrated 18-HEPE, 14-HDHA, and 17-HDHA treatments altered lung levels of specialized pro-resolving lipid mediators (SPMs). 14-HDHA and 17-HDHA treatments more efficiently reduced the exacerbated acute inflammatory response in AgNP exposed MetS mice as compared to 18-HEPE. This included decreased neutrophilic influx, diminished induction of inflammatory cytokines/chemokines, and reduced alterations in SPMs. Examination of SPM receptors determined baseline reductions in MetS mice compared to healthy as well as decreases due to AgNP exposure. Overall, these results demonstrate AgNP exposure disrupts inflammatory resolution, specifically 14-HDHA and 17-HDHA derived SPMs, in MetS contributing to exacerbated acute inflammatory responses. Our findings identify a potential mechanism responsible for enhanced susceptibility in MetS that can be targeted for interventional therapeutic approaches.
Collapse
Affiliation(s)
- Saeed Alqahtani
- School of Health Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN, United States; National Center for Pharmaceuticals, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Li Xia
- School of Health Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN, United States
| | - Amber Jannasch
- Purdue Metabolite Profiling Facility, Purdue University, West Lafayette, IN, United States
| | - Christina Ferreira
- Purdue Metabolite Profiling Facility, Purdue University, West Lafayette, IN, United States
| | - Jackeline Franco
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Jonathan H Shannahan
- School of Health Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN, United States.
| |
Collapse
|
12
|
Zhang J, Li L, Luo L, Yang X, Zhang J, Xie Y, Liang R, Wang W, Lu S. Screening and potential role of tRFs and tiRNAs derived from tRNAs in the carcinogenesis and development of lung adenocarcinoma. Oncol Lett 2021; 22:506. [PMID: 33986867 PMCID: PMC8114470 DOI: 10.3892/ol.2021.12767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Accumulating evidence has indicated that a group of novel molecules, known as transfer RNA (tRNA)-derived fragments (tRFs) and tRNA halves (tiRNAs), which are derived from tRNAs, serve an essential role in numerous types of human disease, in particular solid tumors. However, to the best of our knowledge, the underlying mechanisms of the effect of tRFs and tiRNAs in lung adenocarcinoma have not been reported. The present study aimed to determine the differential expression levels of tRFs and tiRNAs in lung adenocarcinoma and adjacent tissues using a NextSeq system, and further investigated their potential target genes via bioinformatics analysis. Kyoto Encyclopedia of Genes and Genomes signaling pathway and Gene Ontology functional term enrichment analyses were performed to investigate the function of these target genes in the occurrence and development of lung adenocarcinoma. In patients with lung adenocarcinoma, 338 types of tRFs and tiRNAs were detected via sequencing, 284 of which were not previously reported in the tRF database. Compared with the adjacent tissues, 17 types of tRFs and tiRNAs comprising 34 subtypes were found to be abnormally expressed in lung adenocarcinoma tissues, 20 of which were upregulated and 14 downregulated. Reverse transcription-quantitative PCR verification revealed that the expression levels of tiRNA-Lys-CTT-002, tRF-Val-CAC-010 and tRF-Val-CAC-011 were significantly upregulated, while those of tRF-Ser-TGA-005 were downregulated in lung adenocarcinoma tissues. Bioinformatics analysis identified that tRF-Ser-TGA-005 participated in the 'cellular response to transforming growth factor β stimulus' and tRF-Val-CAC-010 and tRF-Val-CAC-011 participated in the 'Hedgehog signaling pathway'. In conclusion, the results of the present study suggested that tRFs and tiRNAs may be closely associated with the pathogenesis and development of lung adenocarcinoma, providing a novel insight for further studies into lung adenocarcinoma.
Collapse
Affiliation(s)
- Jinhua Zhang
- Department of Pathology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Linhui Li
- Department of Pathology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Lilin Luo
- Department of Pathology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Xuantao Yang
- Department of Pathology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Juanjuan Zhang
- Department of Pathology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Yuxin Xie
- Department of Pathology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Rui Liang
- Department of Pathology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Wanpu Wang
- Department of Pathology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Shuaiyao Lu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan 650118, P.R. China
| |
Collapse
|
13
|
Gender specific airway gene expression in COPD sub-phenotypes supports a role of mitochondria and of different types of leukocytes. Sci Rep 2021; 11:12848. [PMID: 34145303 PMCID: PMC8213687 DOI: 10.1038/s41598-021-91742-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 05/20/2021] [Indexed: 01/22/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a destructive inflammatory disease and the genes expressed within the lung are crucial to its pathophysiology. We have determined the RNAseq transcriptome of bronchial brush cells from 312 stringently defined ex-smoker patients. Compared to healthy controls there were for males 40 differentially expressed genes (DEGs) and 73 DEGs for females with only 26 genes shared. The gene ontology (GO) term “response to bacterium” was shared, with several different DEGs contributing in males and females. Strongly upregulated genes TCN1 and CYP1B1 were unique to males and females, respectively. For male emphysema (E)-dominant and airway disease (A)-dominant COPD (defined by computed tomography) the term “response to stress” was found for both sub-phenotypes, but this included distinct up-regulated genes for the E-sub-phenotype (neutrophil-related CSF3R, CXCL1, MNDA) and for the A-sub-phenotype (macrophage-related KLF4, F3, CD36). In E-dominant disease, a cluster of mitochondria-encoded (MT) genes forms a signature, able to identify patients with emphysema features in a confirmation cohort. The MT-CO2 gene is upregulated transcriptionally in bronchial epithelial cells with the copy number essentially unchanged. Both MT-CO2 and the neutrophil chemoattractant CXCL1 are induced by reactive oxygen in bronchial epithelial cells. Of the female DEGs unique for E- and A-dominant COPD, 88% were detected in females only. In E-dominant disease we found a pronounced expression of mast cell-associated DEGs TPSB2, TPSAB1 and CPA3. The differential genes discovered in this study point towards involvement of different types of leukocytes in the E- and A-dominant COPD sub-phenotypes in males and females.
Collapse
|
14
|
Liang ZW, Ge XX, Xu MD, Qin H, Wu MY, Shen M, Zhang Y, Liu XM, Chen K, Li W, Duan W, Qin S. Tumor-associated macrophages promote the metastasis and growth of non-small-cell lung cancer cells through NF-κB/PP2Ac-positive feedback loop. Cancer Sci 2021; 112:2140-2157. [PMID: 33609307 PMCID: PMC8177805 DOI: 10.1111/cas.14863] [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: 09/10/2020] [Revised: 12/29/2020] [Accepted: 01/24/2021] [Indexed: 01/22/2023] Open
Abstract
Non‐small‐cell lung cancer (NSCLC), with its aggressive biological behavior, is one of the most diagnosed cancers. Tumor‐associated inflammatory cells play important roles in the interaction between chronic inflammation and lung cancer, however the mechanisms involved are far from defined. In the present study, by developing an orthotopic NSCLC mouse model based on chronic inflammation, we proved that an inflammatory microenvironment accelerated the growth of orthotopic xenografts in vivo. Tumor‐associated macrophages, the most abundant population of inflammatory cells, were identified. Treatment with macrophage‐conditioned medium (MCM) promoted the growth and migration of NSCLC cells. Using bioinformatics analysis, we identified downregulated PP2Ac expression in NSCLC cells upon treatment with MCM. We further confirmed that this downregulation was executed in an NF‐κB pathway‐dependent manner. As IκB kinase (IKK) has been proved to be a substrate of PP2Ac, inhibition on PP2Ac could result in amplification of NF‐κB pathway signaling. Overexpression of PP2Ac, or the dominant‐negative forms of IKK or IκB, attenuated the acceleration of growth and metastasis by MCM. Using bioinformatics analysis, we further identified that CXCL1 and COL6A1 could be downstream of NF‐κB/PP2Ac pathway. Luciferase assay and ChIP assay further confirmed the location of response elements on the promoter regions of CXCL1 and COL6A1. Elevated CXCL1 facilitated angiogenesis, whereas upregulated COL6A1 promoted proliferation and migration.
Collapse
Affiliation(s)
- Zhan-Wen Liang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin-Xin Ge
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng-Dan Xu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hualong Qin
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng-Yao Wu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Shen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Zhang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiao-Meng Liu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Kai Chen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weiming Duan
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Songbing Qin
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
15
|
Lee S, Kang H, Park D, Yu J, Koh SK, Cho D, Kim D, Kang K, Jeon NL. Modeling 3D Human Tumor Lymphatic Vessel Network Using High‐Throughput Platform. Adv Biol (Weinh) 2021. [DOI: 10.1002/adbi.202000195] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Somin Lee
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Habin Kang
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Dohyun Park
- Department of Mechanical Engineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - James Yu
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Seung Kwon Koh
- Department of Health Sciences and Technology SAIHST Sungkyunkwan University 115, Irwon‐ro, Gangnam‐gu Seoul 06355 Republic of Korea
| | - Duck Cho
- Department of Health Sciences and Technology SAIHST Sungkyunkwan University 115, Irwon‐ro, Gangnam‐gu Seoul 06355 Republic of Korea
- Department of Laboratory Medicine and Genetics Samsung Medical Center Sungkyunkwan University School of Medicine 115, Irwon‐ro, Gangnam‐gu Seoul 06355 Republic of Korea
| | - Da‐Hyun Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Science College of Veterinary Medicine Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Kyung‐Sun Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Science College of Veterinary Medicine Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Noo Li Jeon
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
- Department of Mechanical Engineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
- Institute of Advanced Machinery and Design Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
- Institute of BioEngineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| |
Collapse
|
16
|
Lee J, Lu Y, Oshins R, West J, Moneypenny CG, Han K, Brantly ML. Alpha 1 Antitrypsin-Deficient Macrophages Have Impaired Efferocytosis of Apoptotic Neutrophils. Front Immunol 2020; 11:574410. [PMID: 33329539 PMCID: PMC7714766 DOI: 10.3389/fimmu.2020.574410] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/27/2020] [Indexed: 12/05/2022] Open
Abstract
Alpha 1 antitrypsin deficiency (AATD) is an autosomal co-dominant disorder characterized by a low level of circulating AAT, which significantly reduces protection for the lower airways against proteolytic burden caused by neutrophils. Neutrophils, which are terminally differentiated innate immune cells and play a critical role to clear pathogens, accumulate excessively in the lung of AATD individuals. The neutrophil burden in AATD individuals increases the risk for early-onset destructive lung diseases by producing neutrophil products such as reactive oxygen radicals and various proteases. The level of AAT in AATD individuals is not sufficient to inhibit the activity of neutrophil chemotactic factors such as CXCL-8 and LTB4, which could lead to alveolar neutrophil accumulation in AATD individuals. However, as neutrophils have a short lifespan, and apoptotic neutrophils are rapidly cleared by alveolar macrophages that outnumber the apoptotic neutrophils in the pulmonary alveolus, the increased chemotaxis activity does not fully explain the persistent neutrophil accumulation and the resulting chronic inflammation in AATD individuals. Here, we propose that the ability of alveolar macrophages to clear apoptotic neutrophils is impaired in AATD individuals and it could be the main driver to cause neutrophil accumulation in their lung. This study demonstrates that Z-AAT variant significantly increases the expression of pro-inflammatory cytokines including CXCL-8, CXCL1, LTB4, and TNFα in LPS-treated macrophages. These cytokines play a central role in neutrophil recruitment to the lung and in clearance of apoptotic neutrophils by macrophages. Our result shows that LPS treatment significantly reduces the efferocytosis ability of macrophages with the Z-AAT allele by inducing TNFα expression. We incubated monocyte-derived macrophages (MDMs) with apoptotic neutrophils and found that after 3 h of co-incubation, the expression level of CXCL-8 is reduced in M-MDMs but increased in Z-MDMs. This result shows that the expression of inflammatory cytokines could be increased by impaired efferocytosis. It indicates that the efferocytosis ability of macrophages plays an important role in regulating cytokine expression and resolving inflammation. Findings from this study would help us better understand the multifaceted effect of AAT on regulating neutrophil balance in the lung and the underlying mechanisms.
Collapse
Affiliation(s)
- Jungnam Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Yuanqing Lu
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Regina Oshins
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Jesse West
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Craig G Moneypenny
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Kyudong Han
- Department of Microbiology, College of Science and Technology, Dankook University, Cheonan, South Korea.,DKU-Theragen Institute for NGS Analysis, Cheonan, South Korea
| | - Mark L Brantly
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
17
|
CXCL1 contributes to IL-6 expression in osteoarthritis and rheumatoid arthritis synovial fibroblasts by CXCR2, c-Raf, MAPK, and AP-1 pathway. Arthritis Res Ther 2020; 22:251. [PMID: 33087182 PMCID: PMC7580030 DOI: 10.1186/s13075-020-02331-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/28/2020] [Indexed: 12/26/2022] Open
Abstract
Background Osteoarthritis (OA) and rheumatoid arthritis (RA) are common joint disorders that are considered to be different diseases due to their unique molecular mechanisms and pathogenesis. Chemokines and their corresponding receptors have been well characterized in RA progression, but less so in OA pathogenesis. Methods The human primary synovial fibroblasts (SFs) were obtained from human OA and RA tissue samples. The Western blot and qPCR were performed to analyze the expression levels of CXCL1, as well as CXCL-promoted IL-6 expression in both OASFs and RASFs. The signal cascades that mediate the CXCL1-promoted IL-6 expression were identified by using chemical inhibitors, siRNAs, and shRNAs. Results Here, we found that both diseases feature elevated levels of CXCL1 and interleukin (IL)-6, an important proinflammatory cytokine that participates in OA and RA pathogenesis. In OASFs and RASFs, CXCL1 promoted IL-6 expression in a dose- and time-dependent manner. In OASFs and RASFs overexpressing CXCL1 or transduced with shRNA plasmid, IL-6 expression was markedly upregulated. CXCR2, c-Raf, and MAPKs were found to regulate CXCL1-induced IL-6 expression in OASFs and RASFs. Finally, CXCL1 triggered the transcriptional activities of c-Jun (which regulates the expression of proinflammatory proteins) in OASFs and RASFs. Conclusions Our present work suggests that the CXCL1/CXCR2 axis helps to orchestrate inflammatory responses in OA and RA SFs.
Collapse
|
18
|
Hassan MK, Kumar D, Patel SA, Dixit M. EEF1A2 triggers stronger ERK mediated metastatic program in ER negative breast cancer cells than in ER positive cells. Life Sci 2020; 262:118553. [PMID: 33035587 DOI: 10.1016/j.lfs.2020.118553] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/18/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022]
Abstract
AIMS Ever since EEF1A2's identification as a putative oncogene in breast cancer, it has stimulated curiosity due to its contrasting role in predicting the prognostic values in breast cancer patients. Contradicting reports suggest it to be playing a pro-survival as well as a negative role in the survival of patients. This prompted us to find the association of this protein with molecular subtypes in breast cancer and its effect on EMT in representative cell lines. MAIN METHODS Data-mining was carried out to ascertain the correlation of EEF1A2 with molecular subtypes in breast cancer patients. Scratch wound healing and transwell invasion assays were carried out to assess its role in migration and invasion. Western blot, qRT-PCR, and ELISA were carried out to determine key signalling pathways, cytokines, and EMT factors responsible for the observed phenotype. KEY FINDINGS EEF1A2 was associated with ER receptor positivity in breast cancer and was involved in its transcriptional regulation. It induced a robust metastatic program in MDA-MB-231 (a triple-negative cell line), and induced significant changes in its invasive and migratory properties via activation of the ERK pathway. This was not the case in MCF7 which is an ER-positive cell line. SIGNIFICANCE We highlight the specific tendency of EEF1A2 to enhance invasive properties of cell lines in particular molecular subtype only. This sheds light on its selective role in regulating oncogenic processes in breast cancer and could explain its contradicting association with good survival, despite being an oncogene in a certain cohort of breast cancer patients.
Collapse
Affiliation(s)
- Md Khurshidul Hassan
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, PO-Bhimpur-Padanpur, Jatni, Khurda 752050, Odisha, India
| | - Dinesh Kumar
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, PO-Bhimpur-Padanpur, Jatni, Khurda 752050, Odisha, India
| | - Saket Awadhesbhai Patel
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, PO-Bhimpur-Padanpur, Jatni, Khurda 752050, Odisha, India
| | - Manjusha Dixit
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, PO-Bhimpur-Padanpur, Jatni, Khurda 752050, Odisha, India.
| |
Collapse
|
19
|
Park GY, Pathak HB, Godwin AK, Kwon Y. Epithelial-stromal communication via CXCL1-CXCR2 interaction stimulates growth of ovarian cancer cells through p38 activation. Cell Oncol (Dordr) 2020; 44:77-92. [PMID: 32910411 DOI: 10.1007/s13402-020-00554-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2020] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Paracrine interactions with the stromal environment, including fibroblasts, may be important in the pathogenesis of ovarian cancer. Here, we evaluated the effect of conditioned media derived from ovarian fibroblasts (fibroblast-CMs) and their major cytokines on the growth of ovarian cancer cells, as well as the involvement of mitogen-activated protein kinases (MAPKs) and AKT in mediating this effect. METHODS Ovarian cancer cells were cultured in serum-free media (SF), or conditioned media of fibroblasts derived from normal ovary (CM1) and ovarian tumor tissue (CM2). Cell proliferation was measured by MTT assay. Phosphorylation of MAPKs and AKT was evaluated by Western blotting. Specific inhibitors of MAPKs and AKT were used to evaluate their respective involvement in mediating increased cell growth. Cytokine levels in fibroblast-CMs were measured using Luminex assays. Immunohistochemical staining was conducted for CXCL1, CXCR2 and phosphorylated p38 in primary ovarian tumors. RESULTS CM1 and CM2 significantly increased the growth of ovarian cancer cells relative to SF. In OVCAR3 and OVCAR4 cells, p38 phosphorylation was strongly induced by fibroblast-CMs, and pre-treatment with a p38 inhibitor prevented the growth increase induced by fibroblast-CMs. Fibroblasts secreted high levels of IL-6, IL-8, MCP1 and CXCL1. Treatment with only CXCL1 (1 μg/ml) increased cell growth and p38 phosphorylation. Treatment with a CXCR2 inhibitor effectively prevented p38 activation and cell growth induced by fibroblast-CMs. High expression of both CXCL1 and CXCR2 correlated with high expression of phosphorylated p38 in primary ovarian tumors. CONCLUSIONS From our data, we conclude that CXCL1 is a key factor derived from ovarian fibroblasts that is responsible for increased ovarian cancer cell growth in part through p38 activation. Phosphorylated p38 can be used as a biomarker to predict CXCL1-CXCR2 interaction in vivo.
Collapse
Affiliation(s)
- Geun-Young Park
- Department of Food Science and Engineering, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Harsh B Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA.,University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA.,University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Youngjoo Kwon
- Department of Food Science and Engineering, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea.
| |
Collapse
|
20
|
Mo M, Tong S, Li T, Zu X, Hu X. Serum CXCL13 Level is Associated with Tumor Progression and Unfavorable Prognosis in Penile Cancer. Onco Targets Ther 2020; 13:8757-8769. [PMID: 32943882 PMCID: PMC7473990 DOI: 10.2147/ott.s263980] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/11/2020] [Indexed: 12/28/2022] Open
Abstract
Background Chemokine (C-X-C motif) ligands (CXCLs) are important regulators of tumor progression in many cancers and could serve as potential cancer biomarkers. However, the expression patterns as well as functions of CXCLs remain unclear in penile cancer (PC). The aim of this study was to evaluate the usefulness of serum CXCL13 as a potential cancer biomarker for PC. Patients and Methods This retrospective study enrolled 76 patients diagnosed with PC between 2016 and 2018. Serum CXCL13 level was detected by enzyme-linked immunosorbent assay. Univariable and multivariable Cox regression analyses were conducted to identify the prognostic factors that influence disease-free survival. Human penile cancer cell lines Penl1, Penl2, 149RCa and LM156 were used as in vitro models. The expression of CXCL13 protein in PC cell lines was analyzed by Western blotting. Results Our initial analysis on GSE57955 dataset identified CXCL13 as a top CXCL gene enriched in PC. Higher preoperative serum CXCL13 level was detected in PC cohorts than in healthy male controls (P<0.001). The area under the curve was 0.911 with the sensitivity of 84.2% and specificity of 87.0% to distinguish PC. Preoperative serum CXCL13 level was associated with pathological grade (P=0.048), T stage (P=0.009), nodal status (P<0.001) and pelvic lymph node metastasis (P=0.005) in PC. Serum CXCL13 level could serve as an independent prognostic factor for disease-free survival with a HR of 3.818 (95%CI: 1.126–12.946). Furthermore, autocrine expression of CXCL13 was detected in PC tissues and cell lines. Knockdown of CXCL13 expression suppressed malignant phenotypes (cell proliferation, clonogenesis, apoptosis escape, migration and invasion), attenuated STAT3 and ERK1/2 signaling and reduced MMP2/9 secretion in PC cell lines. Conclusion Serum CXCL13 could serve as a novel diagnostic and prognostic biomarker for PC. CXCL13 signaling might activate oncogenic signaling pathways to promote malignant progression of PC.
Collapse
Affiliation(s)
- Miao Mo
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Shiyu Tong
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Tao Li
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Xiheng Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| |
Collapse
|
21
|
Bernstein DL, Rom S. Let-7g* and miR-98 Reduce Stroke-Induced Production of Proinflammatory Cytokines in Mouse Brain. Front Cell Dev Biol 2020; 8:632. [PMID: 32766248 PMCID: PMC7379105 DOI: 10.3389/fcell.2020.00632] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
Stroke is a debilitating illness facing healthcare today, affecting over 800,000 people and causing over 140,000 deaths each year in the United States. Despite being the third-leading cause of death, very few treatments currently exist for stroke. Often, during an ischemic attack, the blood-brain barrier (BBB) is significantly damaged, which can lead to altered interactions with the immune system, and greatly worsen the damage from a stroke. The impaired, BBB promotes the infiltration of peripheral inflammatory cells into the brain, secreting deleterious mediators (cytokines/chemokines) and resulting in permanent barrier injury. let-7 microRNAs (miRs) are critical for regulating immune responses within the BBB, particularly after ischemic stroke. We have previously shown how transient stroke decreases expression of multiple let-7 miRs, and that restoration of expression confers significant neuroprotection, reduction in brain infiltration by neutrophils, monocytes and T cells. However, the specific mechanisms of action of let-7 miRs remain unexplored, though emerging evidence implicates a range of impacts on cytokines. In the current study, we evaluate the impacts of miR-98 and let-7g* on targeting of cytokine mRNAs, cytokine release following ischemic stroke, and cell-specific changes to the neurovascular space. We determined that miR-98 specifically targets IP-10, while let-7g* specifically aims IL-8, and attenuates their levels. Both produce strong impacts on CCL2 and CCL5. Further, let-7g* strongly improves neurovascular perfusion following ischemic stroke. Together, the results of the study indicate that let-7 miRs are critical for mediating endothelial-immune reactions and improving recovery following ischemic stroke.
Collapse
Affiliation(s)
- David L Bernstein
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Slava Rom
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
22
|
Li Y, Imaizumi T, Matsumiya T, Seya K, Kawaguchi S, Ding J, Ohkuma H. Polyinosinic-Polycytidylic Acid Induces CXCL1 Expression in Cultured hCMEC/D3 Human Cerebral Microvascular Endothelial Cells. Neuroimmunomodulation 2020; 27:38-47. [PMID: 32294654 DOI: 10.1159/000506482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/06/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Brain microvascular endothelial cells are integral components of the blood-brain barrier and play a role in protecting the brain from invading microbes. CXC motif chemokine ligand 1 (CXCL1) induces the chemotaxis of neutrophils, and neutrophils are important in host defense in the brain. However, dysregulated neutrophil infiltration leads to brain diseases. Toll-like receptor 3 (TLR3) is a pattern recognition receptor that recognizes viral double-stranded RNA (dsRNA). The aim of this study was to investigate the effect of an TLR3 agonist on the expression of CXCL1 in brain vascular endothelial cells. METHODS hCMEC/D3 human cerebral microvascular endothelial cells were cultured and treated with polyinosinic-polycytidylic acid (poly IC), a potent synthetic dsRNA agonist for TLR3. The production of CXCL1 mRNA and protein was assessed by real-time RT-PCR and ELISA. The expression of CXCL1 was compared with that of CXCL8. The effect of pretreatment of cells with a NF-κB inhibitor (SN50), a p38 mitogen-activated protein kinase (MAPK) inhibitor (SB203580), a c-Jun N-terminal kinase (JNK) inhibitor (SP600125), an interferon (IFN) regulatory factor 3 inhibitor (MRT67307), and an anti-type I IFN-neutralizing antibody mixture was examined. Phosphorylation of p38 was examined using Western blotting. RESULTS Treating cultured hCMEC/D3 human cells with poly IC induced the expression of CXCL1 as well as another chemokine CXCL8. Pretreatment of cells with SN50, SB203580, and SP600125 decreased the induction of CXCL1 by poly IC. However, it was not affected by MRT67307 or by an anti-type I IFN-neutralizing antibody mixture. Pretreatment of cells with SN50 decreased the poly IC-induced phosphorylation of p38. CONCLUSIONS Poly IC induces the expression of CXCL1 in hCMEC/D3 cells. NF-κB, p38 MAPK, and JNK are involved in this reaction. There is a cross-talk between NF-κB and p38, and NF-κB partially regulates phosphorylation of p38. CXCL1 produced by brain microvascular endothelial cells may contribute to the brain's defense against viral infection and various neurological diseases associated with neutrophil accumulation.
Collapse
Affiliation(s)
- Yuchen Li
- Department of Neurosurgery, Hirosaki University School of Medicine, Hirosaki, Japan
- Department of Vascular Biology, Hirosaki University School of Medicine, Hirosaki, Japan
| | - Tadaatsu Imaizumi
- Department of Vascular Biology, Hirosaki University School of Medicine, Hirosaki, Japan,
| | - Tomoh Matsumiya
- Department of Vascular Biology, Hirosaki University School of Medicine, Hirosaki, Japan
| | - Kazuhiko Seya
- Department of Vascular Biology, Hirosaki University School of Medicine, Hirosaki, Japan
| | - Shogo Kawaguchi
- Department of Vascular Biology, Hirosaki University School of Medicine, Hirosaki, Japan
| | - Jiangli Ding
- Department of Vascular Biology, Hirosaki University School of Medicine, Hirosaki, Japan
| | - Hiroki Ohkuma
- Department of Neurosurgery, Hirosaki University School of Medicine, Hirosaki, Japan
| |
Collapse
|
23
|
Huang L, Chen J, Zhao Y, Gu L, Shao X, Li J, Xu Y, Liu Z, Xu Q. Key candidate genes of STAT1 and CXCL10 in melanoma identified by integrated bioinformatical analysis. IUBMB Life 2019; 71:1634-1644. [PMID: 31216116 DOI: 10.1002/iub.2103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/03/2019] [Indexed: 01/01/2023]
Abstract
The underlying mechanisms and gene signatures of melanoma are unknown. In this study, three expression profile data sets (GSE65568, GSE100050, GSE114445) were integrated to identify candidate genes explaining the pathways and functions of melanoma. Expression data sets including 24 melanoma tumours and 13 normal skin samples were merged and analysed in detail. The three GSE profiles shared 431 differentially expressed genes (DEGs), including 227 upregulated genes, 200 downregulated genes and 4 differentially regulated genes. Moreover, the functions and signalling pathways of the shared DEGs with significant p-values were identified. The two most significant modules were filtered from the DEGs protein-protein interaction (PPI) network, which consisted of 284 nodes. We also plotted the prognostic value of hub genes from an online database. In summary, using integrated bioinformatic analysis, we have identified candidate DEGs and pathways in melanoma that could improve our understanding of the causes and underlying molecular events of melanoma, and these candidate genes and pathways could be therapeutic targets for melanoma.
Collapse
Affiliation(s)
- Lili Huang
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji University Cancer Center, Shanghai, China.,Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, China
| | - Jianhua Chen
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji University Cancer Center, Shanghai, China.,Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, China
| | - Yu Zhao
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji University Cancer Center, Shanghai, China.,Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, China
| | - Linaer Gu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji University Cancer Center, Shanghai, China.,Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, China
| | - Xiaoyan Shao
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji University Cancer Center, Shanghai, China.,Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, China
| | - Jiyu Li
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhuqing Liu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji University Cancer Center, Shanghai, China.,Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, China
| | - Qing Xu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji University Cancer Center, Shanghai, China.,Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, China
| |
Collapse
|
24
|
Luissint AC, Williams HC, Kim W, Flemming S, Azcutia V, Hilgarth RS, Leary MNO, Denning TL, Nusrat A, Parkos CA. Macrophage-dependent neutrophil recruitment is impaired under conditions of increased intestinal permeability in JAM-A-deficient mice. Mucosal Immunol 2019; 12:668-678. [PMID: 30745566 PMCID: PMC6543824 DOI: 10.1038/s41385-019-0143-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023]
Abstract
Junctional adhesion molecule-A (JAM-A) is a transmembrane glycoprotein expressed on leukocytes, endothelia, and epithelia that regulates biological processes including barrier function and immune responses. While JAM-A has been reported to facilitate tissue infiltration of leukocytes under inflammatory conditions, the contributions of leukocyte-expressed JAM-A in vivo remain unresolved. We investigated the role of leukocyte-expressed JAM-A in acute peritonitis induced by zymosan, lipopolysaccharide (LPS), or TNFα using mice with selective loss of JAM-A in myelomonocytic cells (LysM-Cre;Jam-afl/fl). Surprisingly, in LysM-Cre;Jam-afl/fl mice, loss of JAM-A did not affect neutrophil (PMN) recruitment into the peritoneum in response to zymosan, LPS, or TNFα although it was significantly reduced in Jam-aKO mice. In parallel, Jam-aKO peritoneal macrophages exhibited diminished CXCL1 chemokine production and decreased activation of NF-kB, whereas those from LysM-Cre;Jam-afl/fl mice were unaffected. Using Villin-Cre;Jam-afl/fl mice, targeted loss of JAM-A on intestinal epithelial cells resulted in increased intestinal permeability along with reduced peritoneal PMN migration as well as lower levels of CXCL1 and active NF-kB similar to that observed in Jam-aKO animals. Interestingly, in germ-free Villin-Cre;Jam-afl/fl mice, PMN recruitment was unaffected suggesting dependence on gut microbiota. Such observations highlight the functional link between a leaky gut and regulation of innate immune responses.
Collapse
Affiliation(s)
| | - Holly C Williams
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, USA
| | - Wooki Kim
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin, Korea
| | - Sven Flemming
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Veronica Azcutia
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Timothy L Denning
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
Systemic LPS-induced Aβ-solubilization and clearance in AβPP-transgenic mice is diminished by heparanase overexpression. Sci Rep 2019; 9:4600. [PMID: 30872722 PMCID: PMC6418119 DOI: 10.1038/s41598-019-40999-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/26/2019] [Indexed: 12/12/2022] Open
Abstract
Amyloid-β (Aβ) is the main constituent of amyloid deposits in Alzheimer’s disease (AD). The neuropathology is associated with neuroinflammation. Here, we investigated effects of systemic lipopolysaccharide (LPS)-treatment on neuroinflammation and Aβ deposition in AβPP-mice and double-transgenic mice with brain expression of AβPP and heparanase, an enzyme that degrades HS and generates an attenuated LPS-response. At 13 months of age, the mice received a single intraperitoneal injection of 50 µg LPS or vehicle, and were sacrificed 1.5 months thereafter. Aβ in the brain was analyzed histologically and biochemically after sequential detergent extraction. Neuroinflammation was assessed by CD45 immunostaining and mesoscale cytokine/chemokine ELISA. In single-transgenic mice, LPS-treatment reduced total Aβ deposition and increased Tween-soluble Aβ. This was associated with a reduced CXCL1, IL-1β, TNF-α-level and microgliosis, which correlated with amyloid deposition and total Aβ. In contrast, LPS did not change Aβ accumulation or inflammation marker in the double-transgenic mice. Our findings suggest that a single pro-inflammatory LPS-stimulus, if given sufficient time to act, triggers Aβ-clearance in AβPP-transgenic mouse brain. The effects depend on HS and heparanase.
Collapse
|
26
|
Carvalho BC, Oliveira LC, Rocha CD, Fernandes HB, Oliveira IM, Leão FB, Valverde TM, Rego IMG, Ghosh S, Silva AM. Both knock-down and overexpression of Rap2a small GTPase in macrophages result in impairment of NF-κB activity and inflammatory gene expression. Mol Immunol 2019; 109:27-37. [PMID: 30851634 DOI: 10.1016/j.molimm.2019.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 01/12/2019] [Accepted: 02/17/2019] [Indexed: 02/08/2023]
Abstract
Small Ras GTPases are key molecules that regulate a variety of cellular responses in different cell types. Rap1 plays important functions in the regulation of macrophage biology during inflammation triggered by toll-like receptors (TLRs). However, despite sharing a relatively high degree of similarity with Rap1, no studies concerning Rap2 in macrophages and innate immunity have been reported yet. In this work, we show that either way alterations in the levels of Rap2a hampers proper macrophages response to TLR stimulation. Rap2a is activated by LPS in macrophages, and although putative activator TLR-inducible Ras guanine exchange factor RasGEF1b was sufficient to induce, it was not fully required for Rap2a activation. Silencing of Rap2a impaired LPS-induced production of IL-6 cytokine and KC/Cxcl1 chemokine, and also NF-κB activity as measured by reporter gene studies. Surprisingly, overexpression of Rap2a did also lead to marked inhibition of NF-κB activation induced by LPS, Pam3CSK4 and downstream TLR signaling molecules. We also found that Rap2a can inhibit the LPS-induced phosphorylation of the NF-κB subunit p65 at serine 536. Collectively, our data suggest that expression levels of Rap2a in macrophages might be tightly regulated to avoid unbalanced immune response. Our results implicate Rap2a in TLR-mediated responses by contributing to balanced NF-κB activity status in macrophages.
Collapse
Affiliation(s)
- Brener C Carvalho
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Leonardo C Oliveira
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Carolina D Rocha
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Heliana B Fernandes
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Isadora M Oliveira
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Felipe B Leão
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Thalita M Valverde
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Igor M G Rego
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Sankar Ghosh
- Department of Microbiology & Immunology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Aristóbolo M Silva
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| |
Collapse
|
27
|
Liang X, Gupta K, Quintero JR, Cernadas M, Kobzik L, Christou H, Pier GB, Owen CA, Çataltepe S. Macrophage FABP4 is required for neutrophil recruitment and bacterial clearance in Pseudomonas aeruginosa pneumonia. FASEB J 2019; 33:3562-3574. [PMID: 30462529 PMCID: PMC6988858 DOI: 10.1096/fj.201802002r] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/15/2018] [Indexed: 01/29/2023]
Abstract
Fatty acid binding protein 4 (FABP4), an intracellular lipid chaperone and adipokine, is expressed by lung macrophages, but the function of macrophage-FABP4 remains elusive. We investigated the role of FABP4 in host defense in a murine model of Pseudomonas aeruginosa pneumonia. Compared with wild-type (WT) mice, FABP4-deficient (FABP4-/-) mice exhibited decreased bacterial clearance and increased mortality when challenged intranasally with P. aeruginosa. These findings in FABP4-/- mice were associated with a delayed neutrophil recruitment into the lungs and were followed by greater acute lung injury and inflammation. Among leukocytes, only macrophages expressed FABP4 in WT mice with P. aeruginosa pneumonia. Chimeric FABP4-/- mice with WT bone marrow were protected from increased mortality seen in chimeric WT mice with FABP4-/- bone marrow during P. aeruginosa pneumonia, thus confirming the role of macrophages as the main source of protective FABP4 against that infection. There was less production of C-X-C motif chemokine ligand 1 (CXCL1) in FABP4-/- alveolar macrophages and lower airway CXCL1 levels in FABP4-/- mice. Delivering recombinant CXCL1 to the airways protected FABP4-/- mice from increased susceptibility to P. aeruginosa pneumonia. Thus, macrophage-FABP4 has a novel role in pulmonary host defense against P. aeruginosa infection by facilitating crosstalk between macrophages and neutrophils via regulation of macrophage CXCL1 production.-Liang, X., Gupta, K., Rojas Quintero, J., Cernadas, M., Kobzik, L., Christou, H., Pier, G. B., Owen, C. A., Çataltepe, S. Macrophage FABP4 is required for neutrophil recruitment and bacterial clearance in Pseudomonas aeruginosa pneumonia.
Collapse
Affiliation(s)
- Xiaoliang Liang
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kushagra Gupta
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joselyn Rojas Quintero
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Manuela Cernadas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lester Kobzik
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gerald B. Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA; and
| | - Caroline A. Owen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- The Lovelace Respiratory Research Institute, Albuquerque, New Mexico, USA
| | - Sule Çataltepe
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Zonis S, Breunig JJ, Mamelak A, Wawrowsky K, Bresee C, Ginzburg N, Chesnokova V. Inflammation-induced Gro1 triggers senescence in neuronal progenitors: effects of estradiol. J Neuroinflammation 2018; 15:260. [PMID: 30201019 PMCID: PMC6131894 DOI: 10.1186/s12974-018-1298-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/29/2018] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Inflammation has been proposed to contribute to the decline in adult hippocampal neurogenesis. Proinflammatory cytokines activate transcription of chemokine growth-regulated oncogene α (Gro1) in human and murine hippocampal neuronal progenitor cells (NPC). The goal of this study was to investigate the effects of Gro1 on hippocampal neurogenesis in the presence of inflammation. METHODS Human hippocampal NPC were transfected with lentivirus expressing Gro1, and murine NPC and hippocampal neuronal HT-22 cells were treated with Gro1 protein. A plasmid expressing mGro1 was electroporated in the hippocampus of newborn mice that were sacrificed 10 days later. Adult male and female mice were injected with lipopolysaccharide (LPS; 1 mg/kg, i.p in five daily injections) or normal saline. Adult male mice were implanted with pellets releasing 17-β estradiol (E2; 2.5 mg/pellet, 41.666 μg/day release) or placebo for 6 weeks and challenged with LPS or normal saline as above. In both experiments, mice were sacrificed 3 h after the last injection. Hippocampal markers of neurogenesis were assessed in vitro and in vivo by Western blot, real-time PCR, and immunohisto/cytochemistry. RESULTS Gro1 induced premature senescence in NPC and HT-22 cells, activating senescence-associated β-galactosidase and the cell cycle inhibitor p16 and suppressing neuroblast proliferation and expression of doublecortin (DCX) and neuron-specific class III beta-tubulin (Tuj-1), both neuroblast markers, while promoting proliferation of neural glial antigen 2 (Ng2)-positive oligodendrocytes. Gro1 overexpression in the hippocampus of newborn mice resulted in decreased neuroblast development, as evidenced by decreased DCX expression and increased expression of platelet-derived growth factor α receptor (PDGFαR), a marker of oligodendrocyte precursors. In adult mice, Gro1 was induced in response to LPS treatment in male but not in female hippocampus, with a subsequent decrease in neurogenesis and activation of oligodendrocyte progenitors. No changes in neurogenesis were observed in females. Treatment with E2 blunted LPS-induced Gro1 in the male hippocampus. CONCLUSIONS Inflammation-induced Gro1 triggers neuroblast senescence, thus suppressing new neuron development in the hippocampus. Sex-dependent differences in Gro1 response are attributed to estradiol, which blunts these changes, protecting the female hippocampus from the deleterious effects of inflammation-induced Gro1 on neurogenesis.
Collapse
Affiliation(s)
- Svetlana Zonis
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| | - Joshua J. Breunig
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| | - Adam Mamelak
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| | - Kolja Wawrowsky
- Department of Biomedical Science, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| | - Catherine Bresee
- Biostatistics and Bioinformatics Core, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| | - Nadiya Ginzburg
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| | - Vera Chesnokova
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| |
Collapse
|
29
|
Mattingly AJ, Laitano O, Clanton TL. Epinephrine stimulates CXCL1 IL-1 α, IL-6 secretion in isolated mouse limb muscle. Physiol Rep 2018; 5. [PMID: 29192066 PMCID: PMC5727277 DOI: 10.14814/phy2.13519] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 10/27/2017] [Indexed: 12/21/2022] Open
Abstract
Catecholamines stimulate interleukin‐6 (IL‐6) secretion in skeletal muscles. However, whether other cytokines are secreted is currently unknown. Skeletal muscle ex vivo preparations commonly used to study cytokine secretion have dealt with limitations including auto‐oxidation of catecholamines. The use of metal chelators could be an alternative to avoid auto‐oxidation and allow catecholamines to be used at physiological doses. We exposed isolated soleus muscles to 1 or 100 ng/mL epinephrine (EPI) and collected bath samples at 1 and 2 h for multiplex cytokine analysis. Keratinocyte chemoattractant (CXCL1), IL‐6, and IL‐1α were significantly elevated by 100 ng/mL exposure, but not by 1 ng/mL (median [CXCL1] (2 h) = 83 pg/mL; [IL‐6] = 19 pg/mL; IL‐1α = 7.5 pg/mL). CXCL1 and IL‐6 were highly correlated in each sample (P = 0.0001). A second experiment combined the metal chelator, deferoxamine mesylate (DFO), to prevent EPI autoxidation, with 2 ng/mL EPI and 10.5 ng/mL norepinephrine (NOREPI) to mimic peak exercise. Unexpectedly, DFO alone stimulated both IL‐6 and CXCL1 secretion, but together with EPI and NOREPI had no additional effects. Stimulation of cytokine secretory responses from skeletal muscle cells in response to DFO thus precludes its use as a chelating agent in ex vivo models. In conclusion, 100 ng/mL EPI stimulates a robust secretory CXCL1 response, which together with IL‐6 and IL‐1α, may constitute an adrenal‐muscle endocrine response system.
Collapse
Affiliation(s)
- Alex J Mattingly
- Department of Applied Physiology & Kinesiology, University of Florida, College of Health and Human Performance, Gainesville, Florida
| | - Orlando Laitano
- Department of Applied Physiology & Kinesiology, University of Florida, College of Health and Human Performance, Gainesville, Florida
| | - Thomas L Clanton
- Department of Applied Physiology & Kinesiology, University of Florida, College of Health and Human Performance, Gainesville, Florida
| |
Collapse
|
30
|
Zhao W, Erle DJ. Widespread Effects of Chemokine 3' Untranslated Regions on mRNA Degradation and Protein Production in Human Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:1053-1061. [PMID: 29907706 DOI: 10.4049/jimmunol.1800114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/21/2018] [Indexed: 12/16/2022]
Abstract
Chemokines are a large family of chemotactic cytokines that play critical roles in inflammation, development, and diseases. Chemokine expression is highly regulated during development and in response to environmental stimuli. The 3' untranslated regions (3'-UTRs) of mRNA are believed to be important in the control of chemokine gene expression. However, the regulatory effects of most chemokine 3'-UTRs have not been characterized previously. In this work, we systematically studied the effects of 43 CC and CXC chemokine 3'-UTRs on gene expression in eight human cell lines and two types of human primary cells. We found that chemokine 3'-UTRs had a wide spectrum of regulatory effects on mRNA abundance and protein production that were tightly correlated with the effects on mRNA stability. In general, 3'-UTRs had remarkably similar effects across all cell types studied. The presence of AU-rich elements, microRNA targets, and Pumilio binding sites were associated with chemokine 3'-UTR activity but did not fully account for all 3'-UTR activity detected using the reporter assay. Mutational analysis illustrated how specific cis-regulatory elements contributed to the regulatory effect of chemokine 3'-UTRs. These findings bring new insights into the mechanisms by which chemokine expression is regulated by 3'-UTRs.
Collapse
Affiliation(s)
- Wenxue Zhao
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94158; and .,School of Basic Medicine (Shenzhen), Sun Yat-Sen University, Guangzhou 510080, People's Republic of China
| | - David J Erle
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94158; and
| |
Collapse
|
31
|
Tan C, Hu W, He Y, Zhang Y, Zhang G, Xu Y, Tang J. Cytokine-mediated therapeutic resistance in breast cancer. Cytokine 2018; 108:151-159. [PMID: 29609137 DOI: 10.1016/j.cyto.2018.03.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 12/20/2022]
Abstract
Therapeutic resistance leading to tumor relapse is a major challenge in breast cancer (BCa) treatment. Numerous factors involved in multiple mechanisms promote the development of tumor chemo/radio-resistance. Cytokines/chemokines are important inflammatory factors and highly related to tumorigenesis, metastasis and tumors responses to treatment. A large number of studies have demonstrated that the network of cytokines activates multiple cell signaling pathways to promote tumor cell survival, proliferation, invasion, and migration. Particularly in BCa, cytokines-enhanced the epithelial-mesenchymal transition (EMT) process plays a pivotal role in the progression of metastatic phenotypes and resistance to the traditional chemo/radio-therapy. Virtually, therapeutic resistance is not entirely determined by tumor cell intrinsic characteristics but also dependent upon synchronized effects by numerous of local microenvironmental factors. Emerging evidence highlighted that exosomes secreted from various types of cells promote intercellular communication by transferring bioactive molecules including miRNAs and cytokines, suggesting that exosomes are essential for sustentation of tumor progression and therapeutic resistance within the tumor microenvironment. In this review, we discuss the mechanisms by which cytokines promote therapeutic resistance of BCa and suggest a potential approach for improving BCa therapeutics by inhibition of exosome function.
Collapse
Affiliation(s)
- Chunli Tan
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China; School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China; Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, 42 Baiziting, Nanjing 210009, PR China
| | - Weizi Hu
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China; School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China; Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, 42 Baiziting, Nanjing 210009, PR China
| | - Yunjie He
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China
| | - Yanyan Zhang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, 42 Baiziting, Nanjing 210009, PR China
| | - Guangqin Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Yong Xu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, 42 Baiziting, Nanjing 210009, PR China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, 101 Longmian Road, Nanjing 211166, PR China.
| | - Jinhai Tang
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China.
| |
Collapse
|
32
|
West NE, Corrigan JP, Owen RH, Hoole SP, Brown AJ, Blatcher S, Newby AC. Percutaneous Sampling of Local Biomolecule Gradients Across Coronary Artery Atherosclerotic Plaques. JACC Basic Transl Sci 2017; 2:646-654. [PMID: 30062180 PMCID: PMC6058996 DOI: 10.1016/j.jacbts.2017.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 07/04/2017] [Accepted: 07/06/2017] [Indexed: 11/19/2022]
Abstract
A percutaneous catheter device, the Liquid Biopsy System, was developed to sample the unstirred boundary layer of blood upstream and downstream of intact and disrupted human coronary atherosclerotic plaques. Using multiplexed proximity extension assays, release of 20 biomolecules was simultaneously detected in samples taken across plaques before balloon angioplasty, including the soluble form of the endothelial lectin-like oxidized LDL receptor. Additional biomolecules, including matrix metalloproteinase-12, were released after plaque disruption with angioplasty. These experiments demonstrate the power of the Liquid Biopsy System to yield new scientific insights and its ultimate potential to generate new biomarkers and surrogate endpoints for clinical trials.
Collapse
Affiliation(s)
- Nick E.J. West
- Papworth Hospital National Health Service Foundation Trust, Cambridge, United Kingdom
| | | | | | - Stephen P. Hoole
- Papworth Hospital National Health Service Foundation Trust, Cambridge, United Kingdom
| | - Adam J. Brown
- Papworth Hospital National Health Service Foundation Trust, Cambridge, United Kingdom
| | | | - Andrew C. Newby
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
- Address for correspondence: Prof. Andrew C. Newby, British Heart Foundation, Research and Teaching Floor Level 7, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, United Kingdom.
| |
Collapse
|
33
|
Silva RL, Lopes AH, Guimarães RM, Cunha TM. CXCL1/CXCR2 signaling in pathological pain: Role in peripheral and central sensitization. Neurobiol Dis 2017; 105:109-116. [PMID: 28587921 DOI: 10.1016/j.nbd.2017.06.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 02/04/2023] Open
Abstract
Pathological pain conditions can be triggered after peripheral nerve injury and/or inflammation. It is associated with plasticity of nociceptive pathway in which pain is prolonged even after healing of the injured tissue. Generally combinations of analgesic drugs are not sufficient to achieve selective palliation from chronic pain, besides causing a greater number of side effects. In order to identify novel alternatives for more effective treatments, it is necessary to clarify the underlying mechanisms of pathological pain. It is well established that there are two main components in pathological pain development and maintenance: (i) primary sensory neuron sensitization (peripheral sensitization), and (ii) central sensitization. In both components cytokines and chemokines act as key mediators in pain modulation. CXCL1 is a chemokine that promote both nociceptor and central sensitization via its main receptor CXCR2, which is a promising target for novel analgesic drugs. Here, we reviewed and discussed the role of the CXCL1/CXCR2 signaling axis in pathological pain conditions triggered by either peripheral inflammation or nerve injury.
Collapse
Affiliation(s)
- Rangel L Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Alexandre H Lopes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Rafaela M Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; Graduate Program in Basic and Applied Immunology, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
34
|
Morales AJ, Carrero JA, Hung PJ, Tubbs AT, Andrews JM, Edelson BT, Calderon B, Innes CL, Paules RS, Payton JE, Sleckman BP. A type I IFN-dependent DNA damage response regulates the genetic program and inflammasome activation in macrophages. eLife 2017; 6. [PMID: 28362262 PMCID: PMC5409825 DOI: 10.7554/elife.24655] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 03/30/2017] [Indexed: 01/06/2023] Open
Abstract
Macrophages produce genotoxic agents, such as reactive oxygen and nitrogen species, that kill invading pathogens. Here we show that these agents activate the DNA damage response (DDR) kinases ATM and DNA-PKcs through the generation of double stranded breaks (DSBs) in murine macrophage genomic DNA. In contrast to other cell types, initiation of this DDR depends on signaling from the type I interferon receptor. Once activated, ATM and DNA-PKcs regulate a genetic program with diverse immune functions and promote inflammasome activation and the production of IL-1β and IL-18. Indeed, following infection with Listeria monocytogenes, DNA-PKcs-deficient murine macrophages produce reduced levels of IL-18 and are unable to optimally stimulate IFN-γ production by NK cells. Thus, genomic DNA DSBs act as signaling intermediates in murine macrophages, regulating innate immune responses through the initiation of a type I IFN-dependent DDR.
Collapse
Affiliation(s)
- Abigail J Morales
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York City, United States
| | - Javier A Carrero
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States
| | - Putzer J Hung
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States
| | - Anthony T Tubbs
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States
| | - Jared M Andrews
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States
| | - Brian T Edelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States
| | - Boris Calderon
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States
| | - Cynthia L Innes
- Environmental Stress and Cancer Group, National Institute of Environmental Health Sciences, Durham, United States.,NIEHS Microarray Group, National Institute of Environmental Health Sciences, Durham, United States
| | - Richard S Paules
- Environmental Stress and Cancer Group, National Institute of Environmental Health Sciences, Durham, United States.,NIEHS Microarray Group, National Institute of Environmental Health Sciences, Durham, United States
| | - Jacqueline E Payton
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States
| | - Barry P Sleckman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York City, United States
| |
Collapse
|
35
|
Nomiyama H, Osada N, Takahashi I, Terao K, Yamagata K, Yoshie O. Translational Repression of a Splice Variant of Cynomolgus Macaque CXCL1L by Its C-Terminal Sequence. J Interferon Cytokine Res 2017; 37:129-138. [PMID: 28186423 DOI: 10.1089/jir.2016.0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We previously isolated a cDNA clone from cynomolgus macaque encoding a novel CXC chemokine that we termed CXCL1L from its close similarity to CXCL1. However, the cDNA consisted of 3 exons instead of 4 exons that were typically seen in other CXC chemokines. Here, we isolated a cDNA encoding the full-length variant of CXCL1L that we termed CXCL1Lβ. CXCL1Lβ is 50 amino acids longer than the original CXCL1L, which we now term CXCL1Lα. The CXCL1Lβ mRNA is much more abundantly expressed in the cynomolgus macaque tissues than CXCL1Lα mRNA. However, CXCL1Lβ protein was poorly produced by transfected cells compared with that of CXCL1Lα. When the coding region of the fourth exon was fused to the C-terminus of CXCL1 or even to a nonsecretory protein firefly luciferase, the fused proteins were also barely produced, although the mRNAs were abundantly expressed. The polysome profiling analysis suggested that the inhibition was mainly at the translational level. Furthermore, we demonstrated that the C-terminal 5 amino acids of CXCL1Lβ were critical for the translational repression. The present study, thus, reveals a unique translational regulation controlling the production of a splicing variant of CXCL1L. Since the CXCL1L gene is functional only in the Old World monkeys, we also discuss possible reasons for the conservation of the active CXCL1L gene in these monkeys during the primate evolution.
Collapse
Affiliation(s)
- Hisayuki Nomiyama
- 1 Department of Molecular Enzymology, Kumamoto University Graduate School of Medical Sciences , Kumamoto, Japan
| | - Naoki Osada
- 2 Division of Bioengineering and Bioinformatics, Graduate School of Information Science and Technology, Hokkaido University , Sapporo, Japan
| | - Ichiro Takahashi
- 3 Tsukuba Primate Research Center, National Institute of Biomedical Innovation , Health and Nutrition (NIBIOHN), Tsukuba, Japan
| | - Keiji Terao
- 3 Tsukuba Primate Research Center, National Institute of Biomedical Innovation , Health and Nutrition (NIBIOHN), Tsukuba, Japan
| | - Kazuya Yamagata
- 4 Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University , Kumamoto, Japan
| | - Osamu Yoshie
- 5 The Health and Kampo Institute , Sendai, Japan
| |
Collapse
|
36
|
Interleukin-8, CXCL1, and MicroRNA miR-146a Responses to Probiotic Escherichia coli Nissle 1917 and Enteropathogenic E. coli in Human Intestinal Epithelial T84 and Monocytic THP-1 Cells after Apical or Basolateral Infection. Infect Immun 2016; 84:2482-92. [PMID: 27297392 DOI: 10.1128/iai.00402-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/09/2016] [Indexed: 02/08/2023] Open
Abstract
Bacterium-host interactions in the gut proceed via directly contacted epithelial cells, the host's immune system, and a plethora of bacterial factors. Here we characterized and compared exemplary cytokine and microRNA (miRNA) responses of human epithelial and THP-1 cells toward the prototype enteropathogenic Escherichia coli (EPEC) strain E2348/69 (O127:H6) and the probiotic strain Escherichia coli Nissle 1917 (EcN) (O6:K5:H1). Human T84 and THP-1 cells were used as cell culture-based model systems for epithelial and monocytic cells. Polarized T84 monolayers were infected apically or basolaterally. Bacterial challenges from the basolateral side resulted in more pronounced cytokine and miRNA responses than those observed for apical side infections. Interestingly, the probiotic EcN also caused a pronounced transcriptional increase of proinflammatory CXCL1 and interleukin-8 (IL-8) levels when human T84 epithelial cells were infected from the basolateral side. miR-146a, which is known to regulate adaptor molecules in Toll-like receptor (TLR)/NF-κB signaling, was found to be differentially regulated in THP-1 cells between probiotic and pathogenic bacteria. To assess the roles of flagella and flagellin, we employed several flagellin mutants of EcN. EcN flagellin mutants induced reduced IL-8 as well as CXCL1 responses in T84 cells, suggesting that flagellin is an inducer of this cytokine response. Following infection with an EPEC type 3 secretion system (T3SS) mutant, we observed increased IL-8 and CXCL1 transcription in T84 and THP-1 cells compared to that in wild-type EPEC. This study emphasizes the differential induction of miR-146a by pathogenic and probiotic E. coli strains in epithelial and immune cells as well as a loss of probiotic properties in EcN interacting with cells from the basolateral side.
Collapse
|
37
|
Sakai H, Kai Y, Oguchi A, Kimura M, Tabata S, Yaegashi M, Saito T, Sato K, Sato F, Yumoto T, Narita M. Curcumin Inhibits 5-Fluorouracil-induced Up-regulation of CXCL1 and CXCL2 of the Colon Associated with Attenuation of Diarrhoea Development. Basic Clin Pharmacol Toxicol 2016; 119:540-547. [PMID: 27194111 DOI: 10.1111/bcpt.12619] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/03/2016] [Indexed: 12/27/2022]
Abstract
The compound 5-fluorouracil (5-FU) is used in cancer chemotherapy and is known to cause diarrhoea. We recently reported that chemokine (C-X-C motif) ligand 1 (CXCL1) and neutrophils in the colonic mucosa were markedly increased by the administration of 5-FU in mice. Curcumin has anti-inflammatory, antitumour and antioxidant properties. Therefore, we examined the effect of curcumin on 5-FU-induced diarrhoea development and CXCL1 and CXCL2 up-regulation in the colon. Mice were given 5-FU (50 mg/kg, i.p.) daily for 4 days. Curcumin (100 or 300 mg/kg, p.o.) was administered on the day before the first administration of 5-FU and administered 30 min. before the administration of 5-FU. Gene expression levels of CXCL1 and CXCL2 in the colon were examined by real-time RT-PCR. Curcumin reduced the 5-FU-induced diarrhoea development. Under this condition, the CXCL1 and CXCL2 gene up-regulated by 5-FU administration was inhibited by curcumin. The gene expression of CXCL1 and CXCL2 was also enhanced by 5-FU application in vitro. The 5-FU-induced up-regulated CXCL1 and CXCL2 gene expressions were inhibited by curcumin, Bay-117082 and bortezomib, nuclear factor kappa B (NF-κB) inhibitors, C646, a p300/cyclic adenosine monophosphate response element-binding protein-histone acetyltransferase (HAT) inhibitor. In conclusion, these findings suggested that curcumin prevented the development of diarrhoea by inhibiting NF-κB and HAT activation.
Collapse
Affiliation(s)
- Hiroyasu Sakai
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Yuki Kai
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Aya Oguchi
- Department of Pharmacology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Minami Kimura
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Shoko Tabata
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Miyabi Yaegashi
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Taiki Saito
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Ken Sato
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Fumiaki Sato
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Tetsuro Yumoto
- Department of Analytical Pathophysiology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Minoru Narita
- Department of Pharmacology, Hoshi University, Shinagawa-ku, Tokyo, Japan
| |
Collapse
|
38
|
Dileepan M, Sarver AE, Rao SP, Panettieri RA, Subramanian S, Kannan MS. MicroRNA Mediated Chemokine Responses in Human Airway Smooth Muscle Cells. PLoS One 2016; 11:e0150842. [PMID: 26998837 PMCID: PMC4801396 DOI: 10.1371/journal.pone.0150842] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/19/2016] [Indexed: 01/25/2023] Open
Abstract
Airway smooth muscle (ASM) cells play a critical role in the pathophysiology of asthma due to their hypercontractility and their ability to proliferate and secrete inflammatory mediators. microRNAs (miRNAs) are gene regulators that control many signaling pathways and thus serve as potential therapeutic alternatives for many diseases. We have previously shown that miR-708 and miR-140-3p regulate the MAPK and PI3K signaling pathways in human ASM (HASM) cells following TNF-α exposure. In this study, we investigated the regulatory effect of these miRNAs on other asthma-related genes. Microarray analysis using the Illumina platform was performed with total RNA extracted from miR-708 (or control miR)-transfected HASM cells. Inhibition of candidate inflammation-associated gene expression was further validated by qPCR and ELISA. The most significant biologic functions for the differentially expressed gene set included decreased inflammatory response, cytokine expression and signaling. qPCR revealed inhibition of expression of CCL11, CXCL10, CCL2 and CXCL8, while the release of CCL11 was inhibited in miR-708-transfected cells. Transfection of cells with miR-140-3p resulted in inhibition of expression of CCL11, CXCL12, CXCL10, CCL5 and CXCL8 and of TNF-α-induced CXCL12 release. In addition, expression of RARRES2, CD44 and ADAM33, genes known to contribute to the pathophysiology of asthma, were found to be inhibited in miR-708-transfected cells. These results demonstrate that miR-708 and miR-140-3p exert distinct effects on inflammation-associated gene expression and biological function of ASM cells. Targeting these miRNA networks may provide a novel therapeutic mechanism to down-regulate airway inflammation and ASM proliferation in asthma.
Collapse
Affiliation(s)
- Mythili Dileepan
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Anne E. Sarver
- Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Savita P. Rao
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Reynold A. Panettieri
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Subbaya Subramanian
- Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Mathur S. Kannan
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
39
|
Haarberg KMK, Wymore Brand MJ, Overstreet AMC, Hauck CC, Murphy PA, Hostetter JM, Ramer-Tait AE, Wannemuehler MJ. Orally administered extract from Prunella vulgaris attenuates spontaneous colitis in mdr1a -/- mice. World J Gastrointest Pharmacol Ther 2015; 6:223-237. [PMID: 26558156 PMCID: PMC4635162 DOI: 10.4292/wjgpt.v6.i4.223] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/17/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the ability of a Prunella vulgaris (P. vulgaris) ethanolic extract to attenuate spontaneous typhlocolitis in mdr1a-/- mice.
METHODS: Vehicle (5% ethanol) or P. vulgaris ethanolic extract (2.4 mg/d) were administered daily by oral gavage to mdr1a-/- or wild type FVBWT mice from 6 wk of age up to 20 wk of age. Clinical signs of disease were noted by monitoring weight loss. Mice experiencing weight loss in excess of 15% were removed from the study. At the time mice were removed from the study, blood and colon tissue were collected for analyses that included histological evaluation of lesions, inflammatory cytokine levels, and myeloperoxidase activity.
RESULTS: Administration of P. vulgaris extracts to mdr1a-/- mice delayed onset of colitis and reduced severity of mucosal inflammation when compared to vehicle-treated mdr1a-/- mice. Oral administration of the P. vulgaris extract resulted in reduced (P < 0.05) serum levels of IL-10 (4.6 ± 2 vs 19.4 ± 4), CXCL9 (1319.0 ± 277 vs 3901.0 ± 858), and TNFα (9.9 ± 3 vs 14.8 ± 1) as well as reduced gene expression by more than two-fold for Ccl2, Ccl20, Cxcl1, Cxcl9, IL-1α, Mmp10, VCAM-1, ICAM, IL-2, and TNFα in the colonic mucosa of mdr1a-/- mice compared to vehicle-treated mdr1a-/- mice. Histologically, several microscopic parameters were reduced (P < 0.05) in P. vulgaris-treated mdr1a-/- mice, as was myeloperoxidase activity in the colon (2.49 ± 0.16 vs 3.36 ± 0.06, P < 0.05). The numbers of CD4+ T cells (2031.9 ± 412.1 vs 5054.5 ± 809.5) and germinal center B cells (2749.6 ± 473.7 vs 4934.0 ± 645.9) observed in the cecal tonsils of P. vulgaris-treated mdr1a-/- were significantly reduced (P < 0.05) from vehicle-treated mdr1a-/- mice. Vehicle-treated mdr1a-/- mice were found to produce serum antibodies to antigens derived from members of the intestinal microbiota, indicative of severe colitis and a loss of adaptive tolerance to the members of the microbiota. These serum antibodies were greatly reduced or absent in P. vulgaris-treated mdr1a-/- mice.
CONCLUSION: The anti-inflammatory activity of P. vulgaris ethanolic extract effectively attenuated the severity of intestinal inflammation in mdr1a-/- mice.
Collapse
|
40
|
Zou A, Lambert D, Yeh H, Yasukawa K, Behbod F, Fan F, Cheng N. Elevated CXCL1 expression in breast cancer stroma predicts poor prognosis and is inversely associated with expression of TGF-β signaling proteins. BMC Cancer 2014; 14:781. [PMID: 25344051 PMCID: PMC4221705 DOI: 10.1186/1471-2407-14-781] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 10/14/2014] [Indexed: 12/30/2022] Open
Abstract
Background CXCL1 is a chemotactic cytokine shown to regulate breast cancer progression and chemo-resistance. However, the prognostic significance of CXCL1 expression in breast cancer has not been fully characterized. Fibroblasts are important cellular components of the breast tumor microenvironment, and recent studies indicate that this cell type is a potential source of CXCL1 expression in breast tumors. The goal of this study was to further characterize the expression patterns of CXCL1 in breast cancer stroma, determine the prognostic significance of stromal CXCL1 expression, and identify factors affecting stromal CXCL1 expression. Methods Stromal CXCL1 protein expression was analyzed in 54 normal and 83 breast carcinomas by immunohistochemistry staining. RNA expression of CXCL1 in breast cancer stroma was analyzed through data mining in http://www.Oncomine.org. The relationships between CXCL1 expression and prognostic factors were analyzed by univariate analysis. Co-immunofluorescence staining for CXCL1, α-Smooth Muscle Actin (α-SMA) and Fibroblast Specific Protein 1 (FSP1) expression was performed to analyze expression of CXCL1 in fibroblasts. By candidate profiling, the TGF-β signaling pathway was identified as a regulator of CXCL1 expression in fibroblasts. Expression of TGF-β and SMAD gene products were analyzed by immunohistochemistry and data mining analysis. The relationships between stromal CXCL1 and TGF-β signaling components were analyzed by univariate analysis. Carcinoma associated fibroblasts isolated from MMTV-PyVmT mammary tumors were treated with recombinant TGF-β and analyzed for CXCL1 promoter activity by luciferase assay, and protein secretion by ELISA. Results Elevated CXCL1 expression in breast cancer stroma correlated with tumor grade, disease recurrence and decreased patient survival. By co-immunofluorescence staining, CXCL1 expression overlapped with expression of α-SMA and FSP1 proteins. Expression of stromal CXCL1 protein expression inversely correlated with expression of TGF-β signaling components. Treatment of fibroblasts with TGF-β suppressed CXCL1 secretion and promoter activity. Conclusions Increased CXCL1 expression in breast cancer stroma correlates with poor patient prognosis. Furthermore, CXCL1 expression is localized to α-SMA and FSP1 positive fibroblasts, and is negatively regulated by TGF-β signaling. These studies indicate that decreased TGF-β signaling in carcinoma associated fibroblasts enhances CXCL1 expression in fibroblasts, which could contribute to breast cancer progression. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-781) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nikki Cheng
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
41
|
Crotta S, Davidson S, Mahlakoiv T, Desmet CJ, Buckwalter MR, Albert ML, Staeheli P, Wack A. Type I and type III interferons drive redundant amplification loops to induce a transcriptional signature in influenza-infected airway epithelia. PLoS Pathog 2013; 9:e1003773. [PMID: 24278020 PMCID: PMC3836735 DOI: 10.1371/journal.ppat.1003773] [Citation(s) in RCA: 198] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 10/02/2013] [Indexed: 12/20/2022] Open
Abstract
Interferons (IFNs) are a group of cytokines with a well-established antiviral function. They can be induced by viral infection, are secreted and bind to specific receptors on the same or neighbouring cells to activate the expression of hundreds of IFN stimulated genes (ISGs) with antiviral function. Type I IFN has been known for more than half a century. However, more recently, type III IFN (IFNλ, IL-28/29) was shown to play a similar role and to be particularly important at epithelial surfaces. Here we show that airway epithelia, the primary target of influenza A virus, produce both IFN I and III upon infection, and that induction of both depends on the RIG-I/MAVS pathway. While IRF3 is generally regarded as the transcription factor required for initiation of IFN transcription and the so-called "priming loop", we find that IRF3 deficiency has little impact on IFN expression. In contrast, lack of IRF7 reduced IFN production significantly, and only IRF3(-/-)IRF7(-/-) double deficiency completely abolished it. The transcriptional response to influenza infection was largely dependent on IFNs, as it was reduced to a few upregulated genes in epithelia lacking receptors for both type I and III IFN (IFNAR1(-/-)IL-28Rα(-/-)). Wild-type epithelia and epithelia deficient in either the type I IFN receptor or the type III IFN receptor exhibit similar transcriptional profiles in response to virus, indicating that none of the induced genes depends selectively on only one IFN system. In chimeric mice, the lack of both IFN I and III signalling in the stromal compartment alone significantly increased the susceptibility to influenza infection. In conclusion, virus infection of airway epithelia induces, via a RIG-I/MAVS/IRF7 dependent pathway, both type I and III IFNs which drive two completely overlapping and redundant amplification loops to upregulate ISGs and protect from influenza infection.
Collapse
Affiliation(s)
- Stefania Crotta
- Division of Immunoregulation, MRC National Institute for Medical Research, The Ridgeway, London, United Kingdom
| | - Sophia Davidson
- Division of Immunoregulation, MRC National Institute for Medical Research, The Ridgeway, London, United Kingdom
| | - Tanel Mahlakoiv
- Department of Virology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Christophe J. Desmet
- Laboratory of Cellular and Molecular Immunology, Interdisciplinary Cluster of Applied Genoproteomics (GIGA) Research Center and Faculty of Veterinary Medicine, University of Liege, Liege, Belgium
| | - Matthew R. Buckwalter
- Unité Immunobiologie des Cellules Dendritiques, Department of Immunology, Institut Pasteur, Paris, France
- INSERM U818, Paris, France
| | - Matthew L. Albert
- Unité Immunobiologie des Cellules Dendritiques, Department of Immunology, Institut Pasteur, Paris, France
- INSERM U818, Paris, France
- Université Paris Descartes, Paris, France
| | - Peter Staeheli
- Department of Virology, University of Freiburg, Freiburg, Germany
| | - Andreas Wack
- Division of Immunoregulation, MRC National Institute for Medical Research, The Ridgeway, London, United Kingdom
| |
Collapse
|
42
|
Hartman ZC, Poage GM, den Hollander P, Tsimelzon A, Hill J, Panupinthu N, Zhang Y, Mazumdar A, Hilsenbeck SG, Mills GB, Brown PH. Growth of triple-negative breast cancer cells relies upon coordinate autocrine expression of the proinflammatory cytokines IL-6 and IL-8. Cancer Res 2013; 73:3470-80. [PMID: 23633491 PMCID: PMC3853111 DOI: 10.1158/0008-5472.can-12-4524-t] [Citation(s) in RCA: 331] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Triple-negative breast cancers (TNBC) are aggressive with no effective targeted therapies. A combined database analysis identified 32 inflammation-related genes differentially expressed in TNBCs and 10 proved critical for anchorage-independent growth. In TNBC cells, an LPA-LPAR2-EZH2 NF-κB signaling cascade was essential for expression of interleukin (IL)-6, IL-8, and CXCL1. Concurrent inhibition of IL-6 and IL-8 expression dramatically inhibited colony formation and cell survival in vitro and stanched tumor engraftment and growth in vivo. A Cox multivariable analysis of patient specimens revealed that IL-6 and IL-8 expression predicted patient survival times. Together these findings offer a rationale for dual inhibition of IL-6/IL-8 signaling as a therapeutic strategy to improve outcomes for patients with TNBCs.
Collapse
Affiliation(s)
- Zachary C. Hartman
- Department of Clinical Cancer Prevention, The University of Texas - MD Anderson Cancer Center, Houston, TX
| | - Graham M. Poage
- Department of Clinical Cancer Prevention, The University of Texas - MD Anderson Cancer Center, Houston, TX
| | - Petra den Hollander
- Department of Clinical Cancer Prevention, The University of Texas - MD Anderson Cancer Center, Houston, TX
| | - Anna Tsimelzon
- Lester and Sue Smith Breast Center, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Jamal Hill
- Department of Clinical Cancer Prevention, The University of Texas - MD Anderson Cancer Center, Houston, TX
| | | | - Yun Zhang
- Department of Clinical Cancer Prevention, The University of Texas - MD Anderson Cancer Center, Houston, TX
| | - Abhijit Mazumdar
- Department of Clinical Cancer Prevention, The University of Texas - MD Anderson Cancer Center, Houston, TX
| | - Susan G. Hilsenbeck
- Lester and Sue Smith Breast Center, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Gordon B. Mills
- Department of Systems Biology, MD Anderson Cancer Center, Houston, TX
| | - Powel H. Brown
- Department of Clinical Cancer Prevention, The University of Texas - MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
43
|
Rac3 induces a molecular pathway triggering breast cancer cell aggressiveness: differences in MDA-MB-231 and MCF-7 breast cancer cell lines. BMC Cancer 2013; 13:63. [PMID: 23388133 PMCID: PMC3576359 DOI: 10.1186/1471-2407-13-63] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 01/30/2013] [Indexed: 12/19/2022] Open
Abstract
Background Rho GTPases are involved in cellular functions relevant to cancer. The roles of RhoA and Rac1 have already been established. However, the role of Rac3 in cancer aggressiveness is less well understood. Methods This work was conducted to analyze the implication of Rac3 in the aggressiveness of two breast cancer cell lines, MDA-MB-231 and MCF-7: both express Rac3, but MDA-MB-231 expresses more activated RhoA. The effect of Rac3 in cancer cells was also compared with its effect on the non-tumorigenic mammary epithelial cells MCF-10A. We analyzed the consequences of Rac3 depletion by anti-Rac3 siRNA. Results Firstly, we analyzed the effects of Rac3 depletion on the breast cancer cells’ aggressiveness. In the invasive MDA-MB-231 cells, Rac3 inhibition caused a marked reduction of both invasion (40%) and cell adhesion to collagen (84%), accompanied by an increase in TNF-induced apoptosis (72%). This indicates that Rac3 is involved in the cancer cells’ aggressiveness. Secondly, we investigated the effects of Rac3 inhibition on the expression and activation of related signaling molecules, including NF-κB and ERK. Cytokine secretion profiles were also analyzed. In the non-invasive MCF-7 line; Rac3 did not influence any of the parameters of aggressiveness. Conclusions This discrepancy between the effects of Rac3 knockdown in the two cell lines could be explained as follows: in the MDA-MB-231 line, the Rac3-dependent aggressiveness of the cancer cells is due to the Rac3/ERK-2/NF-κB signaling pathway, which is responsible for MMP-9, interleukin-6, -8 and GRO secretion, as well as the resistance to TNF-induced apoptosis, whereas in the MCF-7 line, this pathway is not functional because of the low expression of NF-κB subunits in these cells. Rac3 may be a potent target for inhibiting aggressive breast cancer.
Collapse
|
44
|
A CXCL1 paracrine network links cancer chemoresistance and metastasis. Cell 2012; 150:165-78. [PMID: 22770218 DOI: 10.1016/j.cell.2012.04.042] [Citation(s) in RCA: 852] [Impact Index Per Article: 65.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 01/26/2012] [Accepted: 04/20/2012] [Indexed: 02/07/2023]
Abstract
Metastasis and chemoresistance in cancer are linked phenomena, but the molecular basis for this link is unknown. We uncovered a network of paracrine signals between carcinoma, myeloid, and endothelial cells that drives both processes in breast cancer. Cancer cells that overexpress CXCL1 and 2 by transcriptional hyperactivation or 4q21 amplification are primed for survival in metastatic sites. CXCL1/2 attract CD11b(+)Gr1(+) myeloid cells into the tumor, which produce chemokines including S100A8/9 that enhance cancer cell survival. Although chemotherapeutic agents kill cancer cells, these treatments trigger a parallel stromal reaction leading to TNF-α production by endothelial and other stromal cells. TNF-α via NF-kB heightens the CXCL1/2 expression in cancer cells, thus amplifying the CXCL1/2-S100A8/9 loop and causing chemoresistance. CXCR2 blockers break this cycle, augmenting the efficacy of chemotherapy against breast tumors and particularly against metastasis. This network of endothelial-carcinoma-myeloid signaling interactions provides a mechanism linking chemoresistance and metastasis, with opportunities for intervention.
Collapse
|