1
|
Visuthranukul C, Sriswasdi S, Tepaamorndech S, Chamni S, Leelahavanichkul A, Joyjinda Y, Aksornkitti V, Chomtho S. Enhancing gut microbiota and microbial function with inulin supplementation in children with obesity. Int J Obes (Lond) 2024; 48:1696-1704. [PMID: 39033197 PMCID: PMC11584386 DOI: 10.1038/s41366-024-01590-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/24/2024] [Accepted: 07/10/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND AND OBJECTIVES Gut dysbiosis that resulted from the alteration between host-microbe interaction might worsen obesity-induced systemic inflammation. Gut microbiota manipulation by supplementation of prebiotic inulin may reverse metabolic abnormalities and improve obesity. This study aimed to determine whether inulin supplementation improved intestinal microbiota and microbial functional pathways in children with obesity. METHODS Children with obesity whose BMI above median + 2SDs were recruited to a randomized, double-blinded placebo-controlled study. The participants aged 7-15 years were assigned to inulin supplement extracted from Thai Jerusalem artichoke (intervention), maltodextrin (placebo), and dietary fiber advice groups. All participants received similar monthly conventional advice and follow-up for 6 months. Fecal samples were collected for gut microbiome analysis using 16S rRNA sequencing. Phylogenetic Investigation of Communities by Reconstruction of Unobserved States was performed to infer microbial functional pathways. RESULTS One hundred and forty-three children with available taxonomic and functional pathway abundance profiles were evaluated. A significant increase in alpha-diversity was observed in the inulin group. Inulin supplementation substantially enhanced Bifidobacterium, Blautia, Megasphaera, and several butyrate-producing bacteria, including Agathobacter, Eubacterium coprostanoligenes, and Subdoligranulum, compared to the other groups. The inulin group showed a significant difference in functional pathways of proteasome and riboflavin metabolism. These changes correlated with clinical and metabolic outcomes exclusively in the inulin group. CONCLUSIONS Inulin supplementation significantly promoted gut bacterial diversity and improved gut microbiota dysbiosis in children with obesity. The modulation of functional pathways by inulin suggests its potential to establish beneficial interactions between the gut microbiota and host physiology. Inulin supplementation could be a strategic treatment to restore the balance of intestinal microbiota and regulate their functions in childhood obesity.
Collapse
Affiliation(s)
- Chonnikant Visuthranukul
- Center of Excellence in Pediatric Nutrition, Division of Nutrition, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Sira Sriswasdi
- Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Computational Molecular Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Surapun Tepaamorndech
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Supakarn Chamni
- Center of Excellent in Natural Products and Nanoparticles (NP2), Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Inflammation and Immunology Research Unit (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Yutthana Joyjinda
- WHO-CC for Research and Training on Viral Zoonoses, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Thai Red Cross Emerging Infection Diseases-Health Science Center, Bangkok, 10330, Thailand
| | - Vitavat Aksornkitti
- Center of Excellence in Computational Molecular Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sirinuch Chomtho
- Center of Excellence in Pediatric Nutrition, Division of Nutrition, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
2
|
Shen J, Xue X, Yuan H, Song Y, Wang J, Cui R, Ke K. Deubiquitylating Enzyme OTUB1 Facilitates Neuronal Survival After Intracerebral Hemorrhage Via Inhibiting NF-κB-triggered Apoptotic Cascades. Mol Neurobiol 2024; 61:1726-1736. [PMID: 37775718 DOI: 10.1007/s12035-023-03676-2] [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: 07/04/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023]
Abstract
The deubiquitylase OTU domain-containing ubiquitin aldehyde-binding protein 1 (OTUB1) has been implicated in the pathogenesis of various human diseases. However, the molecular mechanism by which OTUB1 participates in the pathogenesis of intracerebral hemorrhage (ICH) remains elusive. In the present study, we established an autologous whole blood fusion-induced ICH model in C57BL/6 J mice. We showed that the upregulation of OTUB1 contributes to the attenuation of Nuclear factor kappa B (NF-κB) and its downstream apoptotic signaling after ICH. OTUB1 directly associates with NF-κB precursors p105 and p100 after ICH, leading to attenuated polyubiquitylation of p105 and p100. Moreover, we revealed that NF-κB signaling was modestly activated both in ICH tissues and hemin-exposed HT-22 neuronal cells, accompanied with the activation of NF-κB downstream pro-apoptotic signaling. Notably, overexpression of OTUB1 strongly inhibited hemin-induced NF-κB activation, whereas interference of OTUB1 led to the opposite effect. Finally, we revealed that lentiviral transduction of OTUB1 markedly ameliorated hemin-induced apoptotic signaling and HT-22 neuronal death. Collectively, these findings suggest that the upregulation of OTUB1 serves as a neuroprotective mechanism in antagonizing neuroinflammation-induced NF-κB signaling and neuronal death, shed new light on manipulating intracellular deubiquitylating pathways as novel interventive approaches against ICH-induced secondary neuronal damage and death.
Collapse
Affiliation(s)
- Jiabing Shen
- Department of Neurology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, People's Republic of China
| | - Xiaoli Xue
- Department of Neurology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, People's Republic of China
- Department of Neurology, Qidong People's Hospital, Qidong, Jiangsu, People's Republic of China
| | - Huimin Yuan
- Department of Neurology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, People's Republic of China
- Department of Neurology, Qidong People's Hospital, Qidong, Jiangsu, People's Republic of China
| | - Yan Song
- Department of Neurology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, People's Republic of China
| | - Jinglei Wang
- Department of Neurology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, People's Republic of China
- Department of Neurology, Affiliated Hai'an Hospital of Nantong University and Hai'an People's Hospital, Hai'an, People's Republic of China
| | - Ronghui Cui
- Department of Neurology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, People's Republic of China.
| | - Kaifu Ke
- Department of Neurology, Affiliated Hospital and Medical School of Nantong University, Nantong, 226001, People's Republic of China.
| |
Collapse
|
3
|
Upregulated Proteasome Subunits in COVID-19 Patients: A Link with Hypoxemia, Lymphopenia and Inflammation. Biomolecules 2022; 12:biom12030442. [PMID: 35327634 PMCID: PMC8946050 DOI: 10.3390/biom12030442] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 02/01/2023] Open
Abstract
Severe COVID-19 disease leads to hypoxemia, inflammation and lymphopenia. Viral infection induces cellular stress and causes the activation of the innate immune response. The ubiquitin-proteasome system (UPS) is highly implicated in viral immune response regulation. The main function of the proteasome is protein degradation in its active form, which recognises and binds to ubiquitylated proteins. Some proteasome subunits have been reported to be upregulated under hypoxic and hyperinflammatory conditions. Here, we conducted a prospective cohort study of COVID-19 patients (n = 44) and age-and sex-matched controls (n = 20). In this study, we suggested that hypoxia could induce the overexpression of certain genes encoding for subunits from the α and β core of the 20S proteasome and from regulatory particles (19S and 11S) in COVID-19 patients. Furthermore, the gene expression of proteasome subunits was associated with lymphocyte count reduction and positively correlated with inflammatory molecular and clinical markers. Given the importance of the proteasome in maintaining cellular homeostasis, including the regulation of the apoptotic and pyroptotic pathways, these results provide a potential link between COVID-19 complications and proteasome gene expression.
Collapse
|
4
|
Guégan JP, Pollet J, Ginestier C, Charafe-Jauffret E, Peter ME, Legembre P. CD95/Fas suppresses NF-κB activation through recruitment of KPC2 in a CD95L/FasL-independent mechanism. iScience 2021; 24:103538. [PMID: 34917906 PMCID: PMC8666665 DOI: 10.1016/j.isci.2021.103538] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/18/2021] [Accepted: 11/25/2021] [Indexed: 12/28/2022] Open
Abstract
CD95 expression is preserved in triple-negative breast cancers (TNBCs), and CD95 loss in these cells triggers the induction of a pro-inflammatory program, promoting the recruitment of cytotoxic NK cells impairing tumor growth. Herein, we identify a novel interaction partner of CD95, Kip1 ubiquitination-promoting complex protein 2 (KPC2), using an unbiased proteomic approach. Independently of CD95L, CD95/KPC2 interaction contributes to the partial degradation of p105 (NF-κB1) and the subsequent generation of p50 homodimers, which transcriptionally represses NF-κB-driven gene expression. Mechanistically, KPC2 interacts with the C-terminal region of CD95 and serves as an adaptor to recruit RelA (p65) and KPC1, which acts as E3 ubiquitin-protein ligase promoting the degradation of p105 into p50. Loss of CD95 in TNBC cells releases KPC2, limiting the formation of the NF-κB inhibitory homodimer complex (p50/p50), promoting NF-κB activation and the production of pro-inflammatory cytokines, which might contribute to remodeling the immune landscape in TNBC cells.
Collapse
Affiliation(s)
| | - Justine Pollet
- Technological core facility BISCEm, Université de Limoges, US042 Inserm, UMS 2015 CNRS, Centre hospitalo-universitaire de Limoges, Limoges, France
| | - Christophe Ginestier
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Molecular Oncology "Equipe labellisée Ligue Contre le Cancer", Marseille, France
| | - Emmanuelle Charafe-Jauffret
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Molecular Oncology "Equipe labellisée Ligue Contre le Cancer", Marseille, France
| | - Marcus E. Peter
- Division Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Patrick Legembre
- Contrôle de la Réponse Immune B et lymphoproliférations, CRIBL, Université Limoges, UMR CNRS 7276, INSERM 1262, Limoges, France
| |
Collapse
|
5
|
Qi M, Liu X, Zhou Y, Wang H, Zhao Y, Ren J, Xiang J. Berberine Inhibits MDA-MB-231 Cells as an Agonist of G Protein-Coupled Estrogen Receptor 1. Int J Mol Sci 2021; 22:ijms222111466. [PMID: 34768896 PMCID: PMC8583996 DOI: 10.3390/ijms222111466] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 01/18/2023] Open
Abstract
G protein-coupled estrogen receptor 1 (GPER1) is a potential therapeutic target for treating triple-negative breast cancers (TNBC). However, modulators for GPER1 that can be used to treat TNBC have not appeared. Berberine (BBR) is a bioactive isoquinoline alkaloid with high oral safety. In recent years, BBR has shown an inhibitory effect on TNBC tumors such as MDA-MB-231, but the molecular target remains unclear, which hinders related clinical research. Our work proved that BBR is a modulator of GPER1 that can inhibit cell viability, migration, and autophagy of MDA-MB-231 cells. The inhibitory effect of BBR on MDA-MB-231 cells has a dependence on estrogen levels. Although BBR promoted the proteasome, which is a major factor in the degradation of GPER1, it could still induce the protein level of GPER1. Correspondingly, the transcription of cellular communication network factor 2 (CCN2) was promoted. BBR could bind to GPER1 directly and change the secondary structure of GPER1, as in the case of 17β-estradiol (E2). In addition, BBR induced not only a high degree of co-localization of GPER1 and microtubule-associated protein 1 light chain 3 (MAP1LC3), but also the accumulation of sequestosome 1 (SQSTM1/p62) by the inhibition of the nuclear translocation of the nuclear factor-kappa B (NF-κB) subunit (RELA/p65), which indicates NF-κB inhibition and anti-cancer effects. This result proved that the promotional effect of BBR on the GPER1/NF-κB pathway was closely related to its inhibitory effect on autophagy, which may serve as a new mechanism by which to explain the inhibitory effect of BBR on MDA-MB-231 cells and expand our understanding of the function of both BBR and GPER1.
Collapse
Affiliation(s)
- Miaomiao Qi
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (M.Q.); (X.L.); (H.W.); (Y.Z.); (J.R.)
| | - Xiang Liu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (M.Q.); (X.L.); (H.W.); (Y.Z.); (J.R.)
| | - Ying Zhou
- Research Center for Medicine and Structural Biology of Wuhan University, Wuhan University, Wuhan 430071, China;
| | - Haoyu Wang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (M.Q.); (X.L.); (H.W.); (Y.Z.); (J.R.)
| | - Ying Zhao
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (M.Q.); (X.L.); (H.W.); (Y.Z.); (J.R.)
| | - Jing Ren
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (M.Q.); (X.L.); (H.W.); (Y.Z.); (J.R.)
| | - Jin Xiang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (M.Q.); (X.L.); (H.W.); (Y.Z.); (J.R.)
- Correspondence:
| |
Collapse
|
6
|
Claudiani S, Mason CC, Milojkovic D, Bianchi A, Pellegrini C, Di Marco A, Fiol CR, Robinson M, Ponnusamy K, Mokretar K, Chowdhury A, Albert M, Reid AG, Deininger MW, Naresh K, Apperley JF, Khorashad JS. Carfilzomib Enhances the Suppressive Effect of Ruxolitinib in Myelofibrosis. Cancers (Basel) 2021; 13:cancers13194863. [PMID: 34638347 PMCID: PMC8507927 DOI: 10.3390/cancers13194863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022] Open
Abstract
As the first FDA-approved tyrosine kinase inhibitor for treatment of patients with myelofibrosis (MF), ruxolitinib improves clinical symptoms but does not lead to eradication of the disease or significant reduction of the mutated allele burden. The resistance of MF clones against the suppressive action of ruxolitinib may be due to intrinsic or extrinsic mechanisms leading to activity of additional pro-survival genes or signalling pathways that function independently of JAK2/STAT5. To identify alternative therapeutic targets, we applied a pooled-shRNA library targeting ~5000 genes to a JAK2V617F-positive cell line under a variety of conditions, including absence or presence of ruxolitinib and in the presence of a bone marrow microenvironment-like culture medium. We identified several proteasomal gene family members as essential to HEL cell survival. The importance of these genes was validated in MF cells using the proteasomal inhibitor carfilzomib, which also enhanced lethality in combination with ruxolitinib. We also showed that proteasome gene expression is reduced by ruxolitinib in MF CD34+ cells and that additional targeting of proteasomal activity by carfilzomib enhances the inhibitory action of ruxolitinib in vitro. Hence, this study suggests a potential role for proteasome inhibitors in combination with ruxolitinib for management of MF patients.
Collapse
Affiliation(s)
- Simone Claudiani
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Clinton C. Mason
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Utah, Salt Lake City, UT 84108, USA;
| | - Dragana Milojkovic
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Andrea Bianchi
- Department of Information Engineering, University of L’Aquila, 67100 L’Aquila, Italy; (A.B.); (A.D.M.)
| | - Cristina Pellegrini
- Department of Biotechnological and Applied Clinical Science, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Antinisca Di Marco
- Department of Information Engineering, University of L’Aquila, 67100 L’Aquila, Italy; (A.B.); (A.D.M.)
| | - Carme R. Fiol
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Mark Robinson
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Kanagaraju Ponnusamy
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Katya Mokretar
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Avirup Chowdhury
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Michael Albert
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Alistair G. Reid
- Molecular Pathology Unit, Liverpool University, Liverpool L7 8XP, UK;
| | - Michael W. Deininger
- Versiti Blood Research Institute, Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Kikkeri Naresh
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Jane F. Apperley
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Jamshid S. Khorashad
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
- Correspondence:
| |
Collapse
|
7
|
Anti-Inflammatory Activity of 4-((1 R,2 R)-3-Hydroxy-1-(4-hydroxyphenyl)-1-methoxypropan-2-yl)-2-methoxyphenol Isolated from Juglans mandshurica Maxim. in LPS-Stimulated RAW 264.7 Macrophages and Zebrafish Larvae Model. Pharmaceuticals (Basel) 2021; 14:ph14080771. [PMID: 34451869 PMCID: PMC8398860 DOI: 10.3390/ph14080771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022] Open
Abstract
Juglans mandshurica Maxim., a traditional folk medicinal plant, is widely distributed in Korea and China. In our previous study, we isolated a new phenylpropanoid compound, 4-((1R,2R)-3-hydroxy-1-(4-hydroxyphenyl)-1-methoxypropan-2-yl)-2-methoxyphenol (HHMP), from J. mandshurica. In the present study, we evaluated the anti-inflammatory activity of HHMP on lipopolysaccharide (LPS)-stimulated RAW 264.7 cells and zebrafish larvae. HHMP significantly inhibited LPS-induced nitric oxide (NO) and prostaglandin E2 production in a dose-dependent manner. Moreover, HHMP treatment considerably suppressed LPS-induced expression of inducible nitric oxide synthase and cyclooxygenase-2. We also demonstrated the mechanisms of HHMP inhibition of inflammatory responses in LPS-stimulated RAW 264.7 cells via Western blot analysis and immunofluorescence staining. Furthermore, HHMP significantly inhibited NO production in LPS-stimulated zebrafish larvae. Consequently, we established that HHMP significantly inhibited the LPS-induced activation of NF-κB and MAPK and the nuclear translocation of p65 in RAW 264.7 cells. Taken together, our findings demonstrate the effect of HHMP on LPS-induced inflammatory responses in vitro and in vivo, suggesting its potential to be used as a natural anti-inflammatory agent.
Collapse
|
8
|
Mishto M, Rodriguez-Hernandez G, Neefjes J, Urlaub H, Liepe J. Response: Commentary: An In Silico-In Vitro Pipeline Identifying an HLA-A*02:01+ KRAS G12V+ Spliced Epitope Candidate for a Broad Tumor-Immune Response in Cancer Patients. Front Immunol 2021; 12:679836. [PMID: 34326838 PMCID: PMC8315000 DOI: 10.3389/fimmu.2021.679836] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/17/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Michele Mishto
- Centre for Inflammation Biology and Cancer Immunology (CIBCI) & Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom
- Francis Crick Institute, London, United Kingdom
| | - Guillermo Rodriguez-Hernandez
- Centre for Inflammation Biology and Cancer Immunology (CIBCI) & Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom
- Francis Crick Institute, London, United Kingdom
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Henning Urlaub
- Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
- Bioanalytics, Institute of Clinical Chemistry, University Medical Center, Göttingen, Germany
| | - Juliane Liepe
- Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany
| |
Collapse
|
9
|
Gatekeepers of the Gut: The Roles of Proteasomes at the Gastrointestinal Barrier. Biomolecules 2021; 11:biom11070989. [PMID: 34356615 PMCID: PMC8301830 DOI: 10.3390/biom11070989] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022] Open
Abstract
The gut epithelial barrier provides the first line of defense protecting the internal milieu from the environment. To circumvent the exposure to constant challenges such as pathogenic infections and commensal bacteria, epithelial and immune cells at the gut barrier require rapid and efficient means to dynamically sense and respond to stimuli. Numerous studies have highlighted the importance of proteolysis in maintaining homeostasis and adapting to the dynamic changes of the conditions in the gut environment. Primarily, proteolytic activities that are involved in immune regulation and inflammation have been examined in the context of the lysosome and inflammasome activation. Yet, the key to cellular and tissue proteostasis is the ubiquitin–proteasome system, which tightly regulates fundamental aspects of inflammatory signaling and protein quality control to provide rapid responses and protect from the accumulation of proteotoxic damage. In this review, we discuss proteasome-dependent regulation of the gut and highlight the pathophysiological consequences of the disarray of proteasomal control in the gut, in the context of aberrant inflammatory disorders and tumorigenesis.
Collapse
|
10
|
Li W, Liang J, Outeda P, Turner S, Wakimoto BT, Watnick T. A genetic screen in Drosophila reveals an unexpected role for the KIP1 ubiquitination-promoting complex in male fertility. PLoS Genet 2020; 16:e1009217. [PMID: 33378371 PMCID: PMC7802972 DOI: 10.1371/journal.pgen.1009217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 01/12/2021] [Accepted: 10/19/2020] [Indexed: 12/22/2022] Open
Abstract
A unifying feature of polycystin-2 channels is their localization to both primary and motile cilia/flagella. In Drosophila melanogaster, the fly polycystin-2 homologue, Amo, is an ER protein early in sperm development but the protein must ultimately cluster at the flagellar tip in mature sperm to be fully functional. Male flies lacking appropriate Amo localization are sterile due to abnormal sperm motility and failure of sperm storage. We performed a forward genetic screen to identify additional proteins that mediate ciliary trafficking of Amo. Here we report that Drosophila homologues of KPC1 and KPC2, which comprise the mammalian KIP1 ubiquitination-promoting complex (KPC), form a conserved unit that is required for the sperm tail tip localization of Amo. Male flies lacking either KPC1 or KPC2 phenocopy amo mutants and are sterile due to a failure of sperm storage. KPC is a heterodimer composed of KPC1, an E3 ligase, and KPC2 (or UBAC1), an adaptor protein. Like their mammalian counterparts Drosophila KPC1 and KPC2 physically interact and they stabilize one another at the protein level. In flies, KPC2 is monoubiquitinated and phosphorylated and this modified form of the protein is located in mature sperm. Neither KPC1 nor KPC2 directly interact with Amo but they are detected in proximity to Amo at the tip of the sperm flagellum. In summary we have identified a new complex that is involved in male fertility in Drosophila melanogaster.
Collapse
Affiliation(s)
- Weizhe Li
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Jinqing Liang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Patricia Outeda
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Stacey Turner
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Barbara T. Wakimoto
- Department of Biology, University of Washington Seattle, WA, United States of America
| | - Terry Watnick
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States of America
- * E-mail:
| |
Collapse
|
11
|
Chen L, Tian R, Zhang H, Liu X. Anti-mastitis SNV identification of NFκB1 in Chinese Holstein cows and the possible anti-inflammation role of NFκB1/p105 in bovine MECs. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1191-1201. [PMID: 33253378 DOI: 10.1093/abbs/gmaa118] [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: 02/12/2020] [Revised: 07/03/2020] [Accepted: 09/11/2020] [Indexed: 12/29/2022] Open
Abstract
NFκB1/p105 is the critical member of the NFκB family which can suppress inflammation, ageing, and cancer when p50/p50 homodimer is formed. Currently, the research about the role of NFκB1/p105 during cow mastitis is limited. Here, we analyzed the correlation of six single-nucleotide variants of the NFκB1 gene with somatic cell count, milk yield, milk fat content, and milk protein content in 547 Chinese Holstein cows, and explored the mRNA expression profiles of the NFκB family and ubiquitin ligases (βTrCP1, βTrCP2, KPC1, KPC2) in LPS-induced bovine mammary epithelial cells (MECs) by transcriptome-Seq. The association analysis showed that cows with SNV2-TT and SNV6-CC in the NFκB1 gene had significantly higher milk protein content (P < 0.05), while cows with SNV5-TT in the NFκB1 gene had significantly lower somatic cell score (SCS), but CC genotype at SNV5 locus was not detected in our Holstein cows. The transcriptome-Seq results demonstrated the mRNA expression of NFκB1 was increased and peaked at 4 h post-induction, while the mRNA expressions of both KPC1 and BCL3 that promote the anti-inflammation function of NFκB1/p105 were decreased in LPS-induced bovine MECs. TNFAIP3, an inhibitor of both degradation and processing of p105 precursor, was markedly increased by more than 3 folds. Furthermore, bta-miR-125b which targets at the 3'UTR of TNFAIP3 was reduced by 50%. These results indicated that SNV5-TT of the NFκB1 gene with lower SCS may be an anti-mastitis genotype that could cope with infection more efficiently in Chinese Holstein cows. In addition, the anti-inflammation role of NFκB1/p105 seemed to be inhibited in LPS-induced-bovine MECs because the formation of the p50/p50 homodimer was arrested. This study provides a new perspective to understand the inflammatory mechanism in dairy mastitis.
Collapse
Affiliation(s)
- Ling Chen
- School of Modern Agriculture & Biotechnology, Ankang University, Ankang 725000, China
| | - Rongfu Tian
- School of Modern Agriculture & Biotechnology, Ankang University, Ankang 725000, China
| | - Huilin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaolin Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
12
|
Specht G, Roetschke HP, Mansurkhodzhaev A, Henklein P, Textoris-Taube K, Urlaub H, Mishto M, Liepe J. Large database for the analysis and prediction of spliced and non-spliced peptide generation by proteasomes. Sci Data 2020; 7:146. [PMID: 32415162 PMCID: PMC7228940 DOI: 10.1038/s41597-020-0487-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/16/2020] [Indexed: 11/10/2022] Open
Abstract
Proteasomes are the main producers of antigenic peptides presented to CD8+ T cells. They can cut proteins and release their fragments or recombine non-contiguous fragments thereby generating novel sequences, i.e. spliced peptides. Understanding which are the driving forces and the sequence preferences of both reactions can streamline target discovery in immunotherapies against cancer, infection and autoimmunity. Here, we present a large database of spliced and non-spliced peptides generated by proteasomes in vitro, which is available as simple CSV file and as a MySQL database. To generate the database, we performed in vitro digestions of 55 unique synthetic polypeptide substrates with different proteasome isoforms and experimental conditions. We measured the samples using three mass spectrometers, filtered and validated putative peptides, identified 22,333 peptide product sequences (15,028 spliced and 7,305 non-spliced product sequences). Our database and datasets have been deposited to the Mendeley (doi:10.17632/nr7cs764rc.1) and PRIDE (PXD016782) repositories. We anticipate that this unique database can be a valuable source for predictors of proteasome-catalyzed peptide hydrolysis and splicing, with various future translational applications.
Collapse
Affiliation(s)
- Gerd Specht
- Max-Planck-Institute for Biophysical Chemistry, 37077, Göttingen, Germany
| | - Hanna P Roetschke
- Max-Planck-Institute for Biophysical Chemistry, 37077, Göttingen, Germany
| | | | - Petra Henklein
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, D-10117, Berlin, Germany
| | - Kathrin Textoris-Taube
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Shared Facility for Mass Spectrometry, D-10117, Berlin, Germany
| | - Henning Urlaub
- Max-Planck-Institute for Biophysical Chemistry, 37077, Göttingen, Germany
| | - Michele Mishto
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, D-10117, Berlin, Germany.
- Centre for Inflammation Biology and Cancer Immunology (CIBCI) & Peter Gorer Department of Immunobiology, King's College London, SE1 1UL, London, United Kingdom.
| | - Juliane Liepe
- Max-Planck-Institute for Biophysical Chemistry, 37077, Göttingen, Germany.
| |
Collapse
|
13
|
Wang X, Bustos MA, Zhang X, Ramos RI, Tan C, Iida Y, Chang SC, Salomon MP, Tran K, Gentry R, Kravtsova-Ivantsiv Y, Kelly DF, Mills GB, Ciechanover A, Mao Y, Hoon DS. Downregulation of the Ubiquitin-E3 Ligase RNF123 Promotes Upregulation of the NF-κB1 Target SerpinE1 in Aggressive Glioblastoma Tumors. Cancers (Basel) 2020; 12:E1081. [PMID: 32349217 PMCID: PMC7281601 DOI: 10.3390/cancers12051081] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/22/2020] [Accepted: 04/25/2020] [Indexed: 12/28/2022] Open
Abstract
This study examined the role of the ubiquitin E3-ligase RNF123 in modulating downstream NF-κB1 targets in glioblastoma (GB) tumor progression. Our findings revealed an oncogenic pathway (miR-155-5p-RNF123-NF-κB1-p50-SerpinE1) that may represent a new therapeutic target pathway for GB patients with isocitrate dehydrogenase 1 and 2 (IDH) WT (wild type). Mechanistically, we demonstrated that RNF123 is downregulated in IDH WT GB patients and leads to the reduction of p50 levels. RNA-sequencing, reverse-phase protein arrays, and in vitro functional assays on IDH WT GB cell lines with RNF123 overexpression showed that SerpinE1 was a downstream target that is negatively regulated by RNF123. SERPINE1 knockdown reduced the proliferation and invasion of IDH WT GB cell lines. Both SerpinE1 and miR-155-5p overexpression negatively modulated RNF123 expression. In clinical translational analysis, RNF123, SerpinE1, and miR-155-5p were all associated with poor outcomes in GB patients. Multivariable analysis in IDH WT GB patients showed that concurrent low RNF123 and high SerpinE1 was an independent prognostic factor in predicting poor overall survival (p < 0.001, hazard ratio (HR) = 2.93, 95% confidence interval (CI) 1.7-5.05), and an increased risk of recurrence (p < 0.001, relative risk (RR) = 3.56, 95% CI 1.61-7.83).
Collapse
Affiliation(s)
- Xiaowen Wang
- Department of Translational Molecular Medicine, John Wayne Cancer Institute (JWCI) at Providence Saint John’s Health Center, Santa Monica, CA 90404, USA; (X.W.); (M.A.B.); (X.Z.); (R.I.R.); (Y.I.); (M.P.S.); (K.T.); (R.G.); (Y.M.)
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Matias A. Bustos
- Department of Translational Molecular Medicine, John Wayne Cancer Institute (JWCI) at Providence Saint John’s Health Center, Santa Monica, CA 90404, USA; (X.W.); (M.A.B.); (X.Z.); (R.I.R.); (Y.I.); (M.P.S.); (K.T.); (R.G.); (Y.M.)
| | - Xiaoqing Zhang
- Department of Translational Molecular Medicine, John Wayne Cancer Institute (JWCI) at Providence Saint John’s Health Center, Santa Monica, CA 90404, USA; (X.W.); (M.A.B.); (X.Z.); (R.I.R.); (Y.I.); (M.P.S.); (K.T.); (R.G.); (Y.M.)
| | - Romela Irene Ramos
- Department of Translational Molecular Medicine, John Wayne Cancer Institute (JWCI) at Providence Saint John’s Health Center, Santa Monica, CA 90404, USA; (X.W.); (M.A.B.); (X.Z.); (R.I.R.); (Y.I.); (M.P.S.); (K.T.); (R.G.); (Y.M.)
| | - Cong Tan
- Department of Pathology, Cancer Hospital, Fudan University, Shanghai 200032, China;
| | - Yuuki Iida
- Department of Translational Molecular Medicine, John Wayne Cancer Institute (JWCI) at Providence Saint John’s Health Center, Santa Monica, CA 90404, USA; (X.W.); (M.A.B.); (X.Z.); (R.I.R.); (Y.I.); (M.P.S.); (K.T.); (R.G.); (Y.M.)
| | - Shu-Ching Chang
- Medical Data Research Center, Providence Saint Joseph’s Health, Portland, OR 97225, USA;
| | - Matthew P. Salomon
- Department of Translational Molecular Medicine, John Wayne Cancer Institute (JWCI) at Providence Saint John’s Health Center, Santa Monica, CA 90404, USA; (X.W.); (M.A.B.); (X.Z.); (R.I.R.); (Y.I.); (M.P.S.); (K.T.); (R.G.); (Y.M.)
| | - Kevin Tran
- Department of Translational Molecular Medicine, John Wayne Cancer Institute (JWCI) at Providence Saint John’s Health Center, Santa Monica, CA 90404, USA; (X.W.); (M.A.B.); (X.Z.); (R.I.R.); (Y.I.); (M.P.S.); (K.T.); (R.G.); (Y.M.)
| | - Rebecca Gentry
- Department of Translational Molecular Medicine, John Wayne Cancer Institute (JWCI) at Providence Saint John’s Health Center, Santa Monica, CA 90404, USA; (X.W.); (M.A.B.); (X.Z.); (R.I.R.); (Y.I.); (M.P.S.); (K.T.); (R.G.); (Y.M.)
| | - Yelena Kravtsova-Ivantsiv
- The David and Janet Polak Cancer and Vascular Biology Research Center, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Efron Street, Bat-Galim, Haifa 31096, Israel; (Y.K.-I.); (A.C.)
| | - Daniel F. Kelly
- Pacific Neuroscience Institute, JWCI, Santa Monica, CA 90404, USA;
| | - Gordon B. Mills
- Department of Cell Development and Cancer Biology, Knight Cancer Institute, Portland, OR 97239, USA;
| | - Aaron Ciechanover
- The David and Janet Polak Cancer and Vascular Biology Research Center, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Efron Street, Bat-Galim, Haifa 31096, Israel; (Y.K.-I.); (A.C.)
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- The Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Dave S.B. Hoon
- Department of Translational Molecular Medicine, John Wayne Cancer Institute (JWCI) at Providence Saint John’s Health Center, Santa Monica, CA 90404, USA; (X.W.); (M.A.B.); (X.Z.); (R.I.R.); (Y.I.); (M.P.S.); (K.T.); (R.G.); (Y.M.)
| |
Collapse
|
14
|
Arginase Inhibition Supports Survival and Differentiation of Neuronal Precursors in Adult Alzheimer's Disease Mice. Int J Mol Sci 2020; 21:ijms21031133. [PMID: 32046281 PMCID: PMC7037054 DOI: 10.3390/ijms21031133] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 01/23/2023] Open
Abstract
Adult neurogenesis is a complex physiological process, which plays a central role in maintaining cognitive functions, and consists of progenitor cell proliferation, newborn cell migration, and cell maturation. Adult neurogenesis is susceptible to alterations under various physiological and pathological conditions. A substantial decay of neurogenesis has been documented in Alzheimer’s disease (AD) patients and animal AD models; however, several treatment strategies can halt any further decline and even induce neurogenesis. Our previous results indicated a potential effect of arginase inhibition, with norvaline, on various aspects of neurogenesis in triple-transgenic mice. To better evaluate this effect, we chronically administered an arginase inhibitor, norvaline, to triple-transgenic and wild-type mice, and applied an advanced immunohistochemistry approach with several biomarkers and bright-field microscopy. Remarkably, we evidenced a significant reduction in the density of neuronal progenitors, which demonstrate a different phenotype in the hippocampi of triple-transgenic mice as compared to wild-type animals. However, norvaline showed no significant effect upon the progenitor cell number and constitution. We demonstrated that norvaline treatment leads to an escalation of the polysialylated neuronal cell adhesion molecule immunopositivity, which suggests an improvement in the newborn neuron survival rate. Additionally, we identified a significant increase in the hippocampal microtubule-associated protein 2 stain intensity. We also explore the molecular mechanisms underlying the effects of norvaline on adult mice neurogenesis and provide insights into their machinery.
Collapse
|
15
|
NF-kappa B Signaling-Related Signatures Are Connected with the Mesenchymal Phenotype of Circulating Tumor Cells in Non-Metastatic Breast Cancer. Cancers (Basel) 2019; 11:cancers11121961. [PMID: 31817685 PMCID: PMC6966426 DOI: 10.3390/cancers11121961] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/27/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022] Open
Abstract
The role of circulating tumor cells (CTCs), tumor microenvironment (TME), and the immune system in the formation of metastasis is evident, yet the details of their interactions remain unknown. This study aimed at exploring the immunotranscriptome of primary tumors associated with the status of CTCs in breast cancer (BCa) patients. The expression of 730 immune-related genes in formalin-fixed paraffin-embedded samples was analyzed using the multigenomic NanoString technology and correlated with the presence and the phenotype of CTCs. Upregulation of 37 genes and downregulation of 1 gene were observed in patients characterized by a mesenchymal phenotype of CTCs when compared to patients with epithelial CTCs. The upregulated genes were involved in NF-kappa B signaling and in the production of type I interferons. The clinical significance of the differentially expressed genes was evaluated using The Cancer Genome Atlas (TCGA) data of a breast invasive carcinoma (BRCA) cohort. Five of the upregulated genes-PSMD7, C2, IFNAR1, CD84, and CYLD-were independent prognostic factors in terms of overall and disease-free survival. To conclude, our data identify a group of genes that are upregulated in BCa patients with mesenchymal CTCs and reveal their prognostic potential, thus indicating that they merit further investigation.
Collapse
|
16
|
Yuan Y, Wang Y, Liu X, Luo B, Zhang L, Zheng F, Li X, Guo L, Wang L, Jiang M, Pan Y, Yan Y, Yang J, Chen S, Wang J, Tang J. KPC1 alleviates hypoxia/reoxygenation-induced apoptosis in rat cardiomyocyte cells though BAX degradation. J Cell Physiol 2019; 234:22921-22934. [PMID: 31148189 PMCID: PMC6771896 DOI: 10.1002/jcp.28854] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 04/24/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022]
Abstract
Bax triggers cell apoptosis by permeabilizing the outer mitochondrial membrane, leading to membrane potential loss and cytochrome c release. However, it is unclear if proteasomal degradation of Bax is involved in the apoptotic process, especially in heart ischemia-reperfusion (I/R)-induced injury. In the present study, KPC1 expression was heightened in left ventricular cardiomyocytes of patients with coronary heart disease (CHD), in I/R-myocardium in vivo and in hypoxia and reoxygenation (H/R)-induced cardiomyocytes in vitro. Overexpression of KPC1 reduced infarction size and cell apoptosis in I/R rat hearts. Similarly, the forced expression of KPC1 restored mitochondrial membrane potential (MMP) and cytochrome c release driven by H/R in H9c2 cells, whereas reducing cell apoptosis, and knockdown of KPC1 by short-hairpin RNA (shRNA) deteriorated cell apoptosis induced by H/R. Mechanistically, forced expression of KPC1 promoted Bax protein degradation, which was abolished by proteasome inhibitor MG132, suggesting that KPC1 promoted proteasomal degradation of Bax. Furthermore, KPC1 prevented basal and apoptotic stress-induced Bax translocation to mitochondria. Bax can be a novel target for the antiapoptotic effects of KPC1 on I/R-induced cardiomyocyte apoptosis and render mechanistic penetration into at least a subset of the mitochondrial effects of KPC1.
Collapse
Affiliation(s)
- Ye Yuan
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
- Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei ProvinceHubei University of MedicineHubeiChina
| | - Yong‐yi Wang
- Department of Cardiovascular Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xin Liu
- Laboratory Animal CenterHubeiChina
| | - Bin Luo
- Department of Physiology, School of Basic Medicine ScienceHubei University of MedicineHubeiChina
| | - Lei Zhang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
- Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei ProvinceHubei University of MedicineHubeiChina
| | - Fei Zheng
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Xing‐Yuan Li
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Ling‐Yun Guo
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Lu Wang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Miao Jiang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Ya‐mu Pan
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Yu‐wen Yan
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Jian‐ye Yang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Shi‐You Chen
- Department of Physiology & PharmacologyThe University of GeorgiaAthensUSA
| | - Jia‐Ning Wang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
- Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei ProvinceHubei University of MedicineHubeiChina
| | - Jun‐Ming Tang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
- Department of Physiology, School of Basic Medicine ScienceHubei University of MedicineHubeiChina
- Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei ProvinceHubei University of MedicineHubeiChina
| |
Collapse
|
17
|
Yadav D, Mishra BN, Khan F. Quantitative structure-activity relationship and molecular docking studies on human proteasome inhibitors for anticancer activity targeting NF-κB signaling pathway. J Biomol Struct Dyn 2019; 38:3621-3632. [PMID: 31514715 DOI: 10.1080/07391102.2019.1666743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The abnormal ubiquitin-proteasome is found as an important target in various human diseases, especially in cancer, and recently it has received prevalent attention as a challenging therapeutic target. The current work is designed to derive a predictive two-dimensional quantitative structure-activity relationship model for anticancer human proteasome target of NF-κB signaling pathway. The established 2 D-QSAR is dependent on multiple linear regression approach and validated through leave-One-Out and external test set prediction method. The robust QSAR model showed the r2 of 0.83 and q2 of 0.80 and pred_r2 of 0.77. Three chemical properties, electronegativity count, average potential, and T_2_N_6 were identified as significant descriptors to predict the anticancer activities of the proteasome antagonists. Besides, the predicted top hit compounds were considered to check out the compliance with Rule of five and pharmacokinetic parameters for oral bioavailability in the human body. The molecular docking was accomplished to unravel the molecular mode of action of best-predicted compounds which was compatible with the standard drug. Following this approach, lastly two compounds NP and AP were recognized as the best candidates since these top compounds follow all the standard limit point of entire filters and indicated effective and decent docking score. The outcomes of the study sturdily suggested that the developed model and top hit compound's binding conformation are rational in the exploration of unknown antagonist's anticancer activity. The research would be of great support and is supposed to be of immense significance in the development and designing of drug-like candidates in preliminary drug discovery. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Deepika Yadav
- Department of Metabolic and Structural Biology, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, Uttar Pradesh, India
| | - Bhartendu Nath Mishra
- Department of Biotechnology, Institute of Engineering & Technology (a Constituent Institute of Dr. A.P.J. Abdul Kalam Technical University, Lucknow), Lucknow, Uttar Pradesh, India
| | - Feroz Khan
- Department of Metabolic and Structural Biology, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, Uttar Pradesh, India
| |
Collapse
|
18
|
Kocaturk NM, Gozuacik D. Crosstalk Between Mammalian Autophagy and the Ubiquitin-Proteasome System. Front Cell Dev Biol 2018; 6:128. [PMID: 30333975 PMCID: PMC6175981 DOI: 10.3389/fcell.2018.00128] [Citation(s) in RCA: 283] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/13/2018] [Indexed: 12/16/2022] Open
Abstract
Autophagy and the ubiquitin-proteasome system (UPS) are the two major intracellular quality control and recycling mechanisms that are responsible for cellular homeostasis in eukaryotes. Ubiquitylation is utilized as a degradation signal by both systems, yet, different mechanisms are in play. The UPS is responsible for the degradation of short-lived proteins and soluble misfolded proteins whereas autophagy eliminates long-lived proteins, insoluble protein aggregates and even whole organelles (e.g., mitochondria, peroxisomes) and intracellular parasites (e.g., bacteria). Both the UPS and selective autophagy recognize their targets through their ubiquitin tags. In addition to an indirect connection between the two systems through ubiquitylated proteins, recent data indicate the presence of connections and reciprocal regulation mechanisms between these degradation pathways. In this review, we summarize these direct and indirect interactions and crosstalks between autophagy and the UPS, and their implications for cellular stress responses and homeostasis.
Collapse
Affiliation(s)
- Nur Mehpare Kocaturk
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Devrim Gozuacik
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabanci University, Istanbul, Turkey
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul, Turkey
| |
Collapse
|
19
|
Why do proteases mess up with antigen presentation by re-shuffling antigen sequences? Curr Opin Immunol 2018; 52:81-86. [PMID: 29723668 DOI: 10.1016/j.coi.2018.04.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/17/2018] [Indexed: 12/27/2022]
Abstract
The sequence of a large number of MHC-presented epitopes is not present as such in the original antigen because it has been re-shuffled by the proteasome or other proteases. Why do proteases throw a spanner in the works of our model of antigen tagging and immune recognition? We describe in this review what we know about the immunological relevance of post-translationally spliced epitopes and why proteases seem to have a second (dark) personality, which is keen to create new peptide bonds.
Collapse
|
20
|
Li YJ, Zhang GP, Zhao F, Li RQ, Liu SJ, Zhao ZR, Wang X. Target therapy of TRIM-14 inhibits osteosarcoma aggressiveness through the nuclear factor-κB signaling pathway. Exp Ther Med 2017; 15:2365-2373. [PMID: 29467844 PMCID: PMC5792772 DOI: 10.3892/etm.2017.5679] [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] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 05/05/2017] [Indexed: 12/15/2022] Open
Abstract
Osteosarcoma is the most common cause of cancer-associated mortality and the prognosis is yet to be fully elucidated due to the paucity of effective therapeutic targets that significantly influence the quality of life and mean survival rates of patients with osteosarcoma. Studies have showed that tripartite motif-containing (TRIM)-14 is a member of the TRIM protein family that has a vital role in tumor progression and metastasis and promotes angiogenesis, invasion and apoptotic resistance of bone cancer. In this study, a chimeric antibody targeting TRIM-14 (Chanti-TRIM) was constructed and the molecular mechanism of target therapy for TRIM-14 was investigated in osteosarcoma cells and xenograft mice. The growth, migration and invasion properties of U-2OS cells were analyzed following incubation with 10–160 mg/ml Chanti-TRIM. Apoptosis of U-2OS cells was detected after Chanti-TRIM treatment. Matrix metalloproteinase (MMP)-9-mediated nuclear factor-κB (NF-κB) signal pathway was analyzed in U-2OS cells treated with Chanti-TRIM. The inhibitory efficacy of Chanti-TRIM was studied in U-2OS-bearing xenograft mice. Our results demonstrated that neutralizing TRIM-14 expression markedly inhibited the growth, migration and invasion of osteosarcoma cells, in vitro and in vivo. We found that TRIM-14 depletion decreased cell viability and induced cells apoptosis in vitro. In addition, we identified Chanti-TRIM inhibited growth and promoted apoptosis induced by cisplatin through MMP-9-mediated NF-κB signal pathway. Furthermore, we observed that Chanti-TRIM treatment inhibited osteosarcoma growth in vivo. Histological analysis indicated that apoptotic bodies were increased and NF-κB nuclear translocation factors, including Ikkβ, p65 and IkBα, were decreased in tumors treated by Chanti-TRIM. In conclusion, these results showed that Chanti-TRIM markedly inhibited the progression of osteosarcoma, suggesting Chanti-TRIM may be a potential anti-cancer agent that functions via the activation of the NF-κB pathway for osteosarcoma.
Collapse
Affiliation(s)
- Yi-Jiong Li
- Department of Orthopaedics, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Guo-Ping Zhang
- Department of Orthopaedics, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Feng Zhao
- Department of Orthopaedics, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Rui-Qi Li
- Department of Orthopaedics, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Shao-Jun Liu
- Department of Orthopaedics, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Zeng-Ren Zhao
- Department of General Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Xin Wang
- Department of Pathology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| |
Collapse
|