51
|
Wang L, Ni M, Hückelhoven-Krauss A, Sellner L, Hoffmann JM, Neuber B, Luft T, Hegenbart U, Schönland S, Kleist C, Sill M, Chen BA, Wuchter P, Eckstein V, Krüger W, Hilgendorf I, Yerushalmi R, Nagler A, Müller-Tidow C, Ho AD, Dreger P, Schmitt M, Schmitt A. Modulation of B Cells and Homing Marker on NK Cells Through Extracorporeal Photopheresis in Patients With Steroid-Refractory/Resistant Graft-Vs.-Host Disease Without Hampering Anti-viral/Anti-leukemic Effects. Front Immunol 2018; 9:2207. [PMID: 30349527 PMCID: PMC6186805 DOI: 10.3389/fimmu.2018.02207] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/05/2018] [Indexed: 11/13/2022] Open
Abstract
Graft-vs.-host disease (GvHD), a severe complication of allogeneic hematopoietic stem cell transplantation, significantly affects the post-transplant morbidity and mortality. Systemic steroids remain the gold standard for the initial management of GvHD. However, up to 60% of patients will not sufficiently respond to steroids. Extracorporeal photopheresis (ECP), a cell-based immunotherapy, has shown good clinical results in such steroid-refractory/resistant GvHD patients. Given its immunomodulatory, but not global immunosuppressive and steroid-sparing capacity, ECP constitutes an attractive option. In the case of GvHD, the balance of immune cells is destroyed: effector cells are not any longer efficiently controlled by regulatory cells. ECP therapy may restore this balance. However, the precise mechanism and the impact of ECP on anti-viral/anti-leukemic function remain unclear. In this study, 839 ECP treatments were performed on patients with acute GvHD (aGvHD) and chronic GvHD (cGvHD). A comprehensive analysis of effector and regulatory cells in patients under ECP therapy included multi-parametric flow cytometry and tetramer staining, LuminexTM-based cytokine, interferon-γ enzyme-linked immunospot, and chromium-51 release assays. Gene profiling of myeloid-derived suppressor cells (MDSCs) was performed by microarray analysis. Immunologically, modulations of effector and regulatory cells as well as proinflammatory cytokines were observed under ECP treatment: (1) GvHD-relevant cell subsets like CD62L+ NK cells and newly defined CD19hiCD20hi B cells were modulated, but (2) quantity and quality of anti-viral/anti-leukemic effector cells were preserved. (3) The development of MDSCs was promoted and switched from an inactivated subset (CD33-CD11b+) to an activated subset (CD33+CD11b+). (4) The frequency of Foxp3+CD4+ regulatory T cells (Tregs) and CD24+CD38hi regulatory B cells was considerably increased in aGvHD patients, and Foxp3+CD8+ Tregs in cGvHD patients. (5) Proinflammatory cytokines like IL-1β, IL-6, IL-8, and TNF-α were significantly reduced. In summary, ECP constitutes an effective immunomodulatory therapy for patients with steroid-refractory/resistant GvHD without impairment of anti-viral/leukemia effects.
Collapse
Affiliation(s)
- Lei Wang
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Ming Ni
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany.,Department of Hematology, the Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | | | - Leopold Sellner
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Jean-Marc Hoffmann
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Thomas Luft
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Ute Hegenbart
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Stefan Schönland
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Christian Kleist
- Department of Nuclear Medicine, University Clinic Heidelberg, Heidelberg, Germany
| | - Martin Sill
- Division Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - Bao-An Chen
- Department of Hematology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Patrick Wuchter
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany.,German Red Cross Blood Service, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology Mannheim, Mannheim, Germany
| | - Volker Eckstein
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - William Krüger
- Department of Internal Medicine C, Haematology, Oncology, Stem Cell Transplantation, Palliative Care, University Clinic Greifswald, Greifswald, Germany
| | - Inken Hilgendorf
- Department of Internal Medicine II, University Clinic Jena, Jena, Germany
| | - Ronit Yerushalmi
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Anthony D Ho
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| |
Collapse
|
52
|
Kipfer B, Daikeler T, Kuchen S, Hallal M, Andina N, Allam R, Bonadies N. Increased cardiovascular comorbidities in patients with myelodysplastic syndromes and chronic myelomonocytic leukemia presenting with systemic inflammatory and autoimmune manifestations. Semin Hematol 2018; 55:242-247. [DOI: 10.1053/j.seminhematol.2018.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 03/23/2018] [Accepted: 05/06/2018] [Indexed: 12/11/2022]
|
53
|
Singer JW, Fleischman A, Al-Fayoumi S, Mascarenhas JO, Yu Q, Agarwal A. Inhibition of interleukin-1 receptor-associated kinase 1 (IRAK1) as a therapeutic strategy. Oncotarget 2018; 9:33416-33439. [PMID: 30279971 PMCID: PMC6161786 DOI: 10.18632/oncotarget.26058] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 08/15/2018] [Indexed: 02/06/2023] Open
Abstract
Interleukin-1 receptor-associated kinases (IRAK1, IRAK2, IRAK3 [IRAK-M], and IRAK4) are serine-threonine kinases involved in toll-like receptor and interleukin-1 signaling pathways, through which they regulate innate immunity and inflammation. Evidence exists that IRAKs play key roles in the pathophysiologies of cancers, and metabolic and inflammatory diseases, and that IRAK inhibition has potential therapeutic benefits. Molecules capable of selectively interfering with IRAK function and expression have been reported, paving the way for the clinical evaluation of IRAK inhibition. Herein, we focus on IRAK1, review its structure and physiological roles, and summarize emerging data for IRAK1 inhibitors in preclinical and clinical studies.
Collapse
Affiliation(s)
| | - Angela Fleischman
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | | | - John O. Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qiang Yu
- Genome Institute of Singapore, Singapore, SG, Singapore
| | - Anupriya Agarwal
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
54
|
Cheon SY, Kim JM, Kim EJ, Kim SY, Kam EH, Ho CC, Lee SK, Koo BN. Intranuclear delivery of synthetic nuclear factor-kappa B p65 reduces inflammasomes after surgery. Biochem Pharmacol 2018; 158:141-152. [PMID: 30096289 DOI: 10.1016/j.bcp.2018.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023]
Abstract
Patients undergoing surgery can suffer from various complications, including post-operative bleeding, local or systematic infection, and neurologic disorders. Major surgery can initiate innate immune responses and trigger overproduction of inflammatory mediators, which can contribute to organ dysfunction. Inflammasomes are innate immune complexes, which are connected to the pathogenesis of various diseases, including atherosclerosis, hemorrhagic brain injury, and Alzheimer's disease. In the present study, we hypothesized that nucleotide-binding oligomerization domain-containing-like receptor protein (NLRP) inflammasomes may have a role in the pathological effects of surgery. Therefore, we designed a protein inhibitor of nuclear factor kappa B (NF-κB) p65 transcripts, called nt-p65-TMD (nuclear transducible (nt) transcription modulated domain (TMD) of RelA (p65)), that can penetrate the nucleus, and evaluated its therapeutic efficacy for dampening surgery-induced inflammasome activation. It was found that the nt-p65-TMD significantly reduced the NLRP1 inflammasome complex components (NLRP1, ASC, and Caspase-1) and interleukin (IL)-1β and IL-18 productions in the spleen after surgery. In the spleen, specific cell population and selective mediators were altered after surgery with/without nt-p65-TMD treatment. Also, we found that treatment of nt-p65-TMD decreased cell death in the spleen after surgery. Therefore, nt-p65-TMD is a potential novel strategy for reducing surgery-induced NLRP1 inflammasome and complications.
Collapse
Affiliation(s)
- So Yeong Cheon
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeong Min Kim
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Jung Kim
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Yeon Kim
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Hee Kam
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chun-Chang Ho
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Sang-Kyou Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| | - Bon-Nyeo Koo
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
55
|
Mast cells participate in allograft rejection: can IL-37 play an inhibitory role? Inflamm Res 2018; 67:747-755. [PMID: 29961151 DOI: 10.1007/s00011-018-1166-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE The aim of this study was to evaluate the role of mast cells (MCs) in allograft rejection, eventually inhibited by IL-37. Immune cells including MCs participate in allograft rejection by generating IL-1, IL-33, TNF and other cytokines. METHODS We evaluated allograft rejection on the experience of our experimental data and using the relevant literature. RESULTS MCs are involved in initiation and regulation of innate and adaptive immune responses-pathways. MCs are important pro-inflammatory cells which express high-affinity receptor FceRI and can be activated by IgE and some pro-inflammatory cytokines, such as IL-1 and IL-33. The cross-linkage of high affinity IgE receptor on MCs by antigen ligation has a crucial role in allergy, asthma, anaphylaxis, cancer and allograft rejection. MCs mediate immunity in organ transplant, leading to the activation of allospecific T cells implicated in the rejection and generate pro-inflammatory cytokines/chemokines. IL-1 pro-inflammatory cytokine family members released by MCs mediate allograft rejection and inflammation. IL-37 is also an IL-1 family member generated by macrophage cell line in small amounts, which binds to IL-18Rα and produces an anti-inflammatory effect. IL-37 provokes the inhibition of TLR signaling, TLR-induced mTOR and (MyD88)-mediated responses, suppressing pro-inflammatory IL-1 family members and increasing IL-10. CONCLUSION IL-37 inhibition offers the opportunity to immunologically modulate MCs, by suppressing their production of IL-1 family members and reducing the risk of allograft rejection, resulting as a potential good therapeutic new cytokine. Here, we report the relationship between inflammatory MCs, allograft rejection and pro-inflammatory and anti-inflammatory IL-37.
Collapse
|
56
|
Sun X, Zou T, Zuo C, Zhang M, Shi B, Jiang Z, Cui H, Liao X, Li X, Tang Y, Liu Y, Liu X. IL-1α inhibits proliferation and adipogenic differentiation of human adipose-derived mesenchymal stem cells through NF-κB- and ERK1/2-mediated proinflammatory cytokines. Cell Biol Int 2018; 42:794-803. [PMID: 29288588 DOI: 10.1002/cbin.10932] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/26/2017] [Indexed: 12/19/2022]
Abstract
Dysfunctional adipogenesis such as subcutaneous lipoatrophy is closely related to insulin resistance and metabolic disorders. Although the expression or release of the cytokine interleukin-1α (IL-1α) is known to increase in adipose tissue in response to cell death, cell senescence, aging, or solar radiation, the regulatory role of IL-1α in adipogenesis has not been sufficiently investigated. To investigate the problem, we explored the effect of IL-1α on the proliferation and adipogenic differentiation of human adipose-derived mesenchymal stem cells (ADSCs) using cell counting, alamarBlue assay, oil red O staining, Western blot, among others. The results showed that IL-1α evidently inhibited the proliferation and adipogenic differentiation of ADSCs, which might be related with the activated nuclear factor-κB (NF-κB) and extracellular signal-regulated kinase (ERK) 1/2 pathways. Early-stage adipogenic differentiation was more sensitive to IL-1α than late-stage differentiation. After differentiation of ADSCs into mature adipocytes, adding of IL-1α had no obvious influence on the cellular morphology, including lipid droplet accumulation. IL-1α enhanced the expression of proinflammatory cytokines, such as IL-8, IL-6, CCL2 (C-C motif chemokine ligand 2), and IL-1β, when added into the adipogenic medium of ADSCs. Blocking IL-8 and IL-6 with neutralizing antibodies partially alleviated the inhibitory effect of IL-1α on the proliferation and adipogenic differentiation. The results suggest that IL-1α inhibits adipogenesis through activation of NF-κB and ERK1/2 pathways and subsequent upregulation of proinflammatory cytokines in ADSCs. IL-1α might play an important role in mediating lipoatrophy by regulation of ADSCs.
Collapse
Affiliation(s)
- Xuerong Sun
- Institute of Aging Research, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Tangbin Zou
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Changqing Zuo
- Department of Pharmacology, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Mingmeng Zhang
- Institute of Aging Research, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Benyan Shi
- Institute of Aging Research, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Zhiwen Jiang
- Institute of Aging Research, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Hongjing Cui
- Institute of Aging Research, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Xiaoxin Liao
- School of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Xiaoyi Li
- School of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Yuelian Tang
- School of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Yusheng Liu
- Department of Medical Cosmetology, Dongguan People's Hospital, Dongguan, 523059, Guangdong Province, China
| | - Xinguang Liu
- Institute of Aging Research, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China.,Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 5240238, China
| |
Collapse
|
57
|
Targeting JAK2 reduces GVHD and xenograft rejection through regulation of T cell differentiation. Proc Natl Acad Sci U S A 2018; 115:1582-1587. [PMID: 29382747 DOI: 10.1073/pnas.1712452115] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Janus kinase 2 (JAK2) signal transduction is a critical mediator of the immune response. JAK2 is implicated in the onset of graft-versus-host disease (GVHD), which is a significant cause of transplant-related mortality after allogeneic hematopoietic cell transplantation (allo-HCT). Transfer of JAK2-/- donor T cells to allogeneic recipients leads to attenuated GVHD yet maintains graft-versus-leukemia. Th1 differentiation among JAK2-/- T cells is significantly decreased compared with wild-type controls. Conversely, iTreg and Th2 polarization is significantly increased among JAK2-/- T cells. Pacritinib is a multikinase inhibitor with potent activity against JAK2. Pacritinib significantly reduces GVHD and xenogeneic skin graft rejection in distinct rodent models and maintains donor antitumor immunity. Moreover, pacritinib spares iTregs and polarizes Th2 responses as observed among JAK2-/- T cells. Collectively, these data clearly identify JAK2 as a therapeutic target to control donor alloreactivity and promote iTreg responses after allo-HCT or solid organ transplantation. As such, a phase I/II acute GVHD prevention trial combining pacritinib with standard immune suppression after allo-HCT is actively being investigated (https://clinicaltrials.gov/ct2/show/NCT02891603).
Collapse
|
58
|
Sabbione AC, Luna-Vital D, Scilingo A, Añón MC, González de Mejía E. Amaranth peptides decreased the activity and expression of cellular tissue factor on LPS activated THP-1 human monocytes. Food Funct 2018; 9:3823-3834. [DOI: 10.1039/c8fo00323h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The immunomodulatory activity of amaranth peptides is linked for the first time with their antithrombotic activity. Inhibition of tissue factor expression and the NF-κB pathway was observed after treatment with the peptides.
Collapse
Affiliation(s)
- Ana Clara Sabbione
- Food Science and Human Nutrition
- University of Illinois at Urbana-Champaign
- Urbana
- USA
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA)
| | - Diego Luna-Vital
- Food Science and Human Nutrition
- University of Illinois at Urbana-Champaign
- Urbana
- USA
| | - Adriana Scilingo
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA)
- La Plata
- Argentina
| | - María Cristina Añón
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA)
- La Plata
- Argentina
| | | |
Collapse
|
59
|
Zhao X, Bai X, Guan L, Li J, Song X, Ma X, Guo J, Zhang Z, Du Q, Huang Y, Tong D. microRNA-4331 Promotes Transmissible Gastroenteritis Virus (TGEV)-induced Mitochondrial Damage Via Targeting RB1, Upregulating Interleukin-1 Receptor Accessory Protein (IL1RAP), and Activating p38 MAPK Pathway In Vitro. Mol Cell Proteomics 2017; 17:190-204. [PMID: 29217619 PMCID: PMC5795386 DOI: 10.1074/mcp.ra117.000432] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Indexed: 11/06/2022] Open
Abstract
Transmissible gastroenteritis virus (TGEV), a member of the coronaviridae family, could cause fatal diarrhea of piglets and result in numerous economic losses. Previous studies demonstrated that TGEV infection could lead to mitochondrial damage and upregulate miR-4331 level. So miR-4331 may play an important regulatory role in the control of mitochondrial function. To explore the potential role of miR-4331 in mitochondrial damage, we adopted a strategy consisting of quantitative proteomic analysis of porcine kidney (PK-15) cells in response to miR-4331 and TGEV infection. Eventually, 69 differentially expressed proteins were gained. The target of miR-4331 was identified. The effects of miR-4331 and its target RB1 on mitochondrial Ca2+ level, mitochondrial membrane potential (MMP), interleukin-1 receptor accessory protein (IL1RAP), p38 MAPK signaling pathway were investigated. The results showed that miR-4331 elevated mitochondrial Ca2+ level, reduced MMP, targets Retinoblastoma 1 (RB1), upregulated IL1RAP, and induced activation of p38 MAPK pathway during TGEV infection. RB1 was identified as the direct targets of miR-4331 and downregulated IL1RAP, suppressed the activation of p38 MPAK, and attenuated TGEV-induced mitochondrial damage. In addition, IL1RAP played a positive role in activating p38 MAPK signaling and negative role in TGEV-induced mitochondrial damage. The data indicate that miR-4331 aggravates TGEV-induced mitochondrial damage by repressing expression of RB1, promoting IL1RAP, and activating p38 MAPK pathway.
Collapse
Affiliation(s)
- Xiaomin Zhao
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Xiaoyuan Bai
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Lijuan Guan
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Juejun Li
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Xiangjun Song
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Xuelian Ma
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Jianxiong Guo
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Zhichao Zhang
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Qian Du
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Yong Huang
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Dewen Tong
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| |
Collapse
|