1
|
Hong J, Liu W, Xiao X, Gajendran B, Ben-David Y. Targeting pivotal amino acids metabolism for treatment of leukemia. Heliyon 2024; 10:e40492. [PMID: 39654725 PMCID: PMC11626780 DOI: 10.1016/j.heliyon.2024.e40492] [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: 06/27/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024] Open
Abstract
Metabolic reprogramming is a crucial characteristic of cancer, allowing cancer cells to acquire metabolic properties that support their survival, immune evasion, and uncontrolled proliferation. Consequently, targeting cancer metabolism has become an essential therapeutic strategy. Abnormal amino acid metabolism is not only a key aspect of metabolic reprogramming but also plays a significant role in chemotherapy resistance and immune evasion, particularly in leukemia. Changes in amino acid metabolism in tumor cells are typically driven by a combination of signaling pathways and transcription factors. Current approaches to targeting amino acid metabolism in leukemia include inhibiting amino acid transporters, blocking amino acid biosynthesis, and depleting specific amino acids to induce apoptosis in leukemic cells. Different types of leukemic cells rely on the exogenous supply of specific amino acids, such as asparagine, glutamine, arginine, and tryptophan. Therefore, disrupting the supply of these amino acids may represent a vulnerability in leukemia. This review focuses on the pivotal role of amino acids in leukemia metabolism, their impact on leukemic stem cells, and their therapeutic potential.
Collapse
Affiliation(s)
- Jiankun Hong
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| | - Wuling Liu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| | - Xiao Xiao
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| | - Babu Gajendran
- Institute of Pharmacology and Biological Activity, Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, 550014, PR China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550025, Guizhou Province, PR China
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| |
Collapse
|
2
|
Russ E, Mikhalkevich N, Iordanskiy S. Expression of Human Endogenous Retrovirus Group K (HERV-K) HML-2 Correlates with Immune Activation of Macrophages and Type I Interferon Response. Microbiol Spectr 2023; 11:e0443822. [PMID: 36861980 PMCID: PMC10100713 DOI: 10.1128/spectrum.04438-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/30/2023] [Indexed: 03/03/2023] Open
Abstract
Human endogenous retroviruses (HERVs) comprise about 8.3% of the human genome and are capable of producing RNA molecules that can be sensed by pattern recognition receptors, leading to the activation of innate immune response pathways. The HERV-K (HML-2) subgroup is the youngest HERV clade with the highest degree of coding competence. Its expression is associated with inflammation-related diseases. However, the precise HML-2 loci, stimuli, and signaling pathways involved in these associations are not well understood or defined. To elucidate HML-2 expression on a locus-specific level, we used the retroelement sequencing tools TEcount and Telescope to analyze publicly available transcriptome sequencing (RNA-seq) and chromatin immunoprecipitation (ChIP) sequencing data sets of macrophages treated with a wide range of agonists. We found that macrophage polarization significantly correlates with modulation of the expression of specific HML-2 proviral loci. Further analysis demonstrated that the provirus HERV-K102, located in an intergenic region of locus 1q22, constituted the majority of the HML-2 derived transcripts following pro-inflammatory (M1) polarization and was upregulated explicitly in response to interferon gamma (IFN-γ) signaling. We found that signal transducer and activator of transcription 1 and interferon regulatory factor 1 interact with a solo long terminal repeat (LTR) located upstream of HERV-K102, termed LTR12F, following IFN-γ signaling. Using reporter constructs, we demonstrated that LTR12F is critical for HERV-K102 upregulation by IFN-γ. In THP1-derived macrophages, knockdown of HML-2 or knockout of MAVS, an adaptor of RNA-sensing pathways, significantly downregulated genes containing interferon-stimulated response elements (ISREs) in their promoters, suggesting an intermediate role of HERV-K102 in the switch from IFN-γ signaling to the activation of type I interferon expression and, therefore, in a positive feedback loop to enhance pro-inflammatory signaling. IMPORTANCE The human endogenous retrovirus group K subgroup, HML-2, is known to be elevated in a long list of inflammation-associated diseases. However, a clear mechanism for HML-2 upregulation in response to inflammation has not been defined. In this study, we identify a provirus of the HML-2 subgroup, HERV-K102, which is significantly upregulated and constitutes the majority of the HML-2 derived transcripts in response to pro-inflammatory activation of macrophages. Moreover, we identify the mechanism of HERV-K102 upregulation and demonstrate that HML-2 expression enhances interferon-stimulated response element activation. We also demonstrate that this provirus is elevated in vivo and correlates with interferon gamma signaling activity in cutaneous leishmaniasis patients. This study provides key insights into the HML-2 subgroup and suggests that it may participate in enhancing pro-inflammatory signaling in macrophages and probably other immune cells.
Collapse
Affiliation(s)
- Eric Russ
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- Graduate Program of Cellular and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Natallia Mikhalkevich
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Sergey Iordanskiy
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Wu Z, Han T, Su H, Xuan J, Wang X. Comprehensive analysis of fatty acid and lactate metabolism–related genes for prognosis value, immune infiltration, and therapy in osteosarcoma patients. Front Oncol 2022; 12:934080. [PMID: 36119478 PMCID: PMC9478861 DOI: 10.3389/fonc.2022.934080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Osteosarcoma is the most frequent bone tumor. Notwithstanding that significant medical progress has been achieved in recent years, the 5-year overall survival of osteosarcoma patients is inferior. Regulation of fatty acids and lactate plays an essential role in cancer metabolism. Therefore, our study aimed to comprehensively assess the fatty acid and lactate metabolism pattern and construct a fatty acid and lactate metabolism–related risk score system to predict prognosis in osteosarcoma patients. Clinical data and RNA expression data were downloaded from the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) and Gene Expression Omnibus (GEO) databases. We used the least absolute shrinkage and selection operator (LASSO) and Cox regression analyses to construct a prognostic risk score model. Relationships between the risk score model and age, gender, tumor microenvironment characteristics, and drug sensitivity were also explored by correlation analysis. We determined the expression levels of prognostic genes in osteosarcoma cells via Western blotting. We developed an unknown fatty acid and lactate metabolism–related risk score system based on three fatty acid and lactate metabolism–related genes (SLC7A7, MYC, and ACSS2). Survival analysis showed that osteosarcoma patients in the low-risk group were likely to have a better survival time than those in the high-risk group. The area under the curve (AUC) value shows that our risk score model performs well in predicting prognosis. Elevated fatty acids and lactate risk scores weaken immune function and the environment of the body, which causes osteosarcoma patients’ poor survival outcomes. In general, the constructed fatty acid and lactate metabolism–related risk score model can offer essential insights into subsequent mechanisms in available research. In addition, our study may provide rational treatment strategies for clinicians based on immune correlation analysis and drug sensitivity in the future.
Collapse
Affiliation(s)
- Zhouwei Wu
- Department of Orthopaedic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tao Han
- Department of Orthopaedic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haohan Su
- Department of Orthopaedic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiangwei Xuan
- Department of Orthopaedic Surgery, Zhuji Affiliated Hospital of Wenzhou Medical University, Zhuji, China
- *Correspondence: Xinwei Wang, ; Jiangwei Xuan,
| | - Xinwei Wang
- Department of Orthopaedic Surgery, Zhuji Affiliated Hospital of Wenzhou Medical University, Zhuji, China
- *Correspondence: Xinwei Wang, ; Jiangwei Xuan,
| |
Collapse
|
4
|
Liao P, Chang N, Xu B, Qiu Y, Wang S, Zhou L, He Y, Xie X, Li Y. Amino acid metabolism: challenges and opportunities for the therapeutic treatment of leukemia and lymphoma. Immunol Cell Biol 2022; 100:507-528. [PMID: 35578380 DOI: 10.1111/imcb.12557] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 02/23/2022] [Accepted: 05/14/2022] [Indexed: 11/26/2022]
Abstract
Leukemia and lymphoma-the most common hematological malignant diseases-are often accompanied by complications such as drug resistance, refractory diseases and relapse. Amino acids (AAs) are important energy sources for malignant cells. Tumor-mediated AA metabolism is associated with the immunosuppressive properties of the tumor microenvironment, thereby assisting malignant cells to evade immune surveillance. Targeting abnormal AA metabolism in the tumor microenvironment may be an effective therapeutic approach to address the therapeutic challenges of leukemia and lymphoma. Here, we review the effects of glutamine, arginine and tryptophan metabolism on tumorigenesis and immunomodulation, and define the differences between tumor cells and immune effector cells. We also comment on treatments targeting these AA metabolism pathways in lymphoma and leukemia and discuss how these treatments have profound adverse effects on tumor cells, but leave the immune cells unaffected or mildly affected.
Collapse
Affiliation(s)
- Peiyun Liao
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ning Chang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Binyan Xu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yingqi Qiu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Sheng Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lijuan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanjie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoling Xie
- Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
5
|
Rotoli BM, Visigalli R, Ferrari F, Ranieri M, Tamma G, Dall’Asta V, Barilli A. Desmopressin Stimulates Nitric Oxide Production in Human Lung Microvascular Endothelial Cells. Biomolecules 2022; 12:biom12030389. [PMID: 35327581 PMCID: PMC8945551 DOI: 10.3390/biom12030389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 01/10/2023] Open
Abstract
Desmopressin (dDAVP) is the best characterized analogue of vasopressin, the endocrine regulator of water balance endowed with potent vasoconstrictive effects. Despite the use of dDAVP in clinical practice, ranging from the treatment of nephrogenic diabetes insipidus to bleeding disorders, much remains to be understood about the impact of the drug on endothelial phenotype. The aim of this study was, thus, to evaluate the effects of desmopressin on the viability and function of human pulmonary microvascular endothelial cells (HLMVECs). The results obtained demonstrate that the vasopressor had no cytotoxic effect on the endothelium; similarly, no sign of endothelial activation was induced by dDAVP, indicated by the lack of effect on the expression of inflammatory cytokines and adhesion molecules. Conversely, the drug significantly stimulated the production of nitric oxide (NO) and the expression of the inducible isoform of nitric oxide synthase, NOS2/iNOS. Since the intracellular level of cAMP also increased, we can hypothesize that NO release is consequent to the activation of the vasopressin receptor 2 (V2R)/guanylate cyclase (Gs)/cAMP axis. Given the multifaceted role of NOS2-deriving NO for many physio-pathological conditions, the meanings of these findings in HLMVECs appears intriguing and deserves to be further addressed.
Collapse
Affiliation(s)
- Bianca Maria Rotoli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (B.M.R.); (R.V.); (F.F.); (V.D.)
| | - Rossana Visigalli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (B.M.R.); (R.V.); (F.F.); (V.D.)
| | - Francesca Ferrari
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (B.M.R.); (R.V.); (F.F.); (V.D.)
| | - Marianna Ranieri
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (M.R.); (G.T.)
| | - Grazia Tamma
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (M.R.); (G.T.)
| | - Valeria Dall’Asta
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (B.M.R.); (R.V.); (F.F.); (V.D.)
| | - Amelia Barilli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (B.M.R.); (R.V.); (F.F.); (V.D.)
- Correspondence:
| |
Collapse
|
6
|
Sheng L, Luo Q, Chen L. Amino Acid Solute Carrier Transporters in Inflammation and Autoimmunity. Drug Metab Dispos 2022; 50:DMD-AR-2021-000705. [PMID: 35152203 DOI: 10.1124/dmd.121.000705] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/14/2022] [Accepted: 01/27/2022] [Indexed: 02/21/2024] Open
Abstract
The past decade exposed the importance of many homeostasis and metabolism related proteins in autoimmunity disease and inflammation. Solute carriers (SLCs) are a group of membrane channels that can transport amino acids, the building blocks of proteins, nutrients, and neurotransmitters. This review summarizes the role of SLCs amino acid transporters in inflammation and autoimmunity disease. In detail, the importance of Glutamate transporters SLC1A1, SLC1A2, and SLC1A3, mainly expressed in the brain where they help prevent glutamate excitotoxicity, is discussed in the context of central nervous system disorders such as multiple sclerosis. Similarly, the cationic amino acid transporter SLC7A1 (CAT1), which is an important arginine transporter for T cells, and SLC7A2 (CAT2), essential for innate immunity. SLC3 family proteins, which bind with light chains from the SLC7 family (SLC7A5, SLC7A7 and SLC7A11) to form heteromeric amino acid transporters, are also explored to describe their roles in T cells, NK cells, macrophages and tumor immunotherapies. Altogether, the link between SLC amino acid transporters with inflammation and autoimmunity may contribute to a better understanding of underlying mechanism of disease and provide novel potential therapeutic avenues. Significance Statement SIGNIFICANCE STATEMENT In this review, we summarize the link between SLC amino acid transporters and inflammation and immune responses, specially SLC1 family members and SLC7 members. Studying the link may contribute to a better understanding of related diseases and provide potential therapeutic targets and useful to the researchers who have interest in the involvement of amino acids in immunity.
Collapse
Affiliation(s)
| | - Qi Luo
- Tsinghua University, China
| | | |
Collapse
|
7
|
Barilli A, Visigalli R, Ferrari F, Di Lascia M, Riccardi B, Puccini P, Dall'Asta V, Rotoli BM. Organic cation transporters (OCTs/OCTNs) in human primary alveolar epithelial cells. Biochem Biophys Res Commun 2021; 576:27-32. [PMID: 34478916 DOI: 10.1016/j.bbrc.2021.08.076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/25/2021] [Indexed: 12/23/2022]
Abstract
Alveolar epithelium, besides exerting a key role in gas exchange and surfactant production, plays important functions in host defense and inflammation. Pathological conditions associated to alveolar dysfunction include Acute Respiratory Distress Syndrome (ARDS), asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). The use of predictive in vitro models of human alveolar epithelium is nowadays required for the study of disease mechanisms, as well as of pharmacokinetic parameters of pulmonary drugs delivery. Here, we employed a novel 3D model of human alveoli, namely EpiAlveolar™, consisting of primary alveolar epithelial cells, pulmonary endothelial cells and fibroblasts, that reflects properly the in vivo-like conditions. In EpiAlveolar™ we performed a characterization of Organic Cation Transporters (OCTs and OCTNs) expression and activity and we found that OCTN2, OCT1 and OCT3 are expressed on the basolateral membrane; instead, ATB0,+ transporter for cationic and neutral amino acids, which shares with OCTN2 the affinity for carnitine as substrate, is readily detectable and functional at the apical side. We also show that these transporters differentially interact with anticholinergic drugs. Overall, our findings reveal close similarities of EpiAlveolar™ with the tracheal/bronchial epithelium (EpiAirway™ model) and entrust this alveolar tissue as a potential tool for the screening of biopharmaceuticals molecules.
Collapse
Affiliation(s)
- Amelia Barilli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125, Parma, Italy
| | - Rossana Visigalli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125, Parma, Italy
| | - Francesca Ferrari
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125, Parma, Italy
| | - Maria Di Lascia
- Preclinical Pharmacokinetics, Biochemistry & Metabolism Dept., Chiesi Farmaceutici, Largo F. Belloli 11/A, 43122, Parma, Italy
| | - Benedetta Riccardi
- Preclinical Pharmacokinetics, Biochemistry & Metabolism Dept., Chiesi Farmaceutici, Largo F. Belloli 11/A, 43122, Parma, Italy
| | - Paola Puccini
- Preclinical Pharmacokinetics, Biochemistry & Metabolism Dept., Chiesi Farmaceutici, Largo F. Belloli 11/A, 43122, Parma, Italy
| | - Valeria Dall'Asta
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125, Parma, Italy.
| | - Bianca Maria Rotoli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125, Parma, Italy
| |
Collapse
|
8
|
Dai W, Feng J, Hu X, Chen Y, Gu Q, Gong W, Feng T, Wu J. SLC7A7 is a prognostic biomarker correlated with immune infiltrates in non-small cell lung cancer. Cancer Cell Int 2021; 21:106. [PMID: 33632211 PMCID: PMC7905560 DOI: 10.1186/s12935-021-01781-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 01/20/2021] [Indexed: 12/14/2022] Open
Abstract
Background SLC7A7 (solute carrier family 7, amino acid transporter light chain, y + L system, member 7) is a critical gene in the regulation of cationic amino acid transport. However, the relationships between SLC7A7 and prognosis and tumor-infiltrating lymphocytes in different cancers remain unclear. Methods SLC7A7 expression was analyzed using the Oncomine database and Tumor Immune Estimation Resource (TIMER) site. The enrichment of the GO (Gene Oncology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways was conducted by DAVID. We evaluated the influence of SLC7A7 on clinical prognosis using the PrognoScan database. The functional state of SLC7A7 in various types of cancers was analyzed by CancerSEA. The relationships between SLC7A7 and cancer immune infiltrates was investigated by TIMER. Furthermore, correlations between SLC7A7 expression and gene marker sets of immune infiltrates were analyzed by TIMER and Gene Expression Profiling Interactive Analysis (GEPIA). The expression of SLC7A7 was verified by GEO database and immunohistochemistry. Results A lung cancer cohort study (GSE31210) showed that high SLC7A7 expression was associated with poor overall survival (OS) and relapse-free survival (RFS). In addition, SLC7A7 had a significant impact on the prognosis of diverse cancers. SLC7A7 expression was positively correlated with infiltrating levels of CD4 + and CD8 + T cells, macrophages, neutrophils and dendritic cells (DCs) in non-small cell lung cancer (NSCLC). SLC7A7 expression was also strongly correlated with various immune marker sets in NSCLC. Conclusions These results indicated a role for SLC7A7 in infiltration of CD8 + T cells, CD4 + T cells, tumor-associated macrophages (TAMs), neutrophils and DCs in multiple cancers, and regulation of T cell exhaustion and Tregs in NSCLC. These findings suggest that SLC7A7 could be served as a biomarker for prognosis and immune infiltration in NSCLC.
Collapse
Affiliation(s)
- Wumin Dai
- Research center, Cancer Hospital of University of Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Jianguo Feng
- Research center, Cancer Hospital of University of Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Xiao Hu
- Department of Abdominal Oncology, Cancer Hospital of University of Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Yongyi Chen
- Clinical Laboratory, Cancer Hospital of University of Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Qing Gu
- Department of Abdominal Oncology, Cancer Hospital of University of Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Wangang Gong
- Research center, Cancer Hospital of University of Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Tingting Feng
- Department of Thoracic Oncology Radiotherapy, Cancer Hospital of University of Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Jie Wu
- Clinical Laboratory, Cancer Hospital of University of Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| |
Collapse
|
9
|
Mikhalkevich N, O’Carroll IP, Tkavc R, Lund K, Sukumar G, Dalgard CL, Johnson KR, Li W, Wang T, Nath A, Iordanskiy S. Response of human macrophages to gamma radiation is mediated via expression of endogenous retroviruses. PLoS Pathog 2021; 17:e1009305. [PMID: 33556144 PMCID: PMC7895352 DOI: 10.1371/journal.ppat.1009305] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/19/2021] [Accepted: 01/11/2021] [Indexed: 01/11/2023] Open
Abstract
Ionizing radiation-induced tissue damage recruits monocytes into the exposed area where they are differentiated to macrophages. These implement phagocytic removal of dying cells and elicit an acute inflammatory response, but can also facilitate tumorigenesis due to production of anti-inflammatory cytokines. Using primary human monocyte-derived macrophages (MDMs) and the THP1 monocytic cell line, we demonstrate that gamma radiation triggers monocyte differentiation toward the macrophage phenotype with increased expression of type I interferons (IFN-I) and both pro- and anti-inflammatory macrophage activation markers. We found that these changes correlate with significantly upregulated expression of 622 retroelements from various groups, particularly of several clades of human endogenous retroviruses (HERVs). Elevated transcription was detected in both sense and antisense directions in the HERV subgroups tested, including the most genetically homogeneous clade HML-2. The level of antisense transcription was three- to five-fold higher than of the sense strand levels. Using a proximity ligation assay and immunoprecipitation followed by RNA quantification, we identified an increased amount of the dsRNA receptors MDA-5 and TLR3 bound to an equivalent number of copies of sense and antisense chains of HERVK HML-2 RNA. This binding triggered MAVS-associated signaling pathways resulting in increased expression of IFN-I and inflammation related genes that enhanced the cumulative inflammatory effect of radiation-induced senescence. HML-2 knockdown was accompanied with reduced expression and secretion of IFNα, pro-inflammatory (IL-1β, IL-6, CCL2, CCL3, CCL8, and CCL20) and anti-inflammatory (IL10) modulators in irradiated monocytes and MDMs. Taken together, our data indicate that radiation stress-induced HERV expression enhances the IFN-I and cytokine response and results in increased levels of pro-inflammatory modulators along with expression of anti-inflammatory factors associated with the macrophage tumorigenic phenotype. Ionizing radiation is a powerful stressogenic factor that induces massive cell damage. The signals released from radiation-damaged tissues recruit the monocytes, which are differentiated into macrophages that remove dying cells via phagocytosis and facilitate inflammation but can also contribute to tumorigenesis through anti-inflammatory and regenerative activities. The mechanism of this dual response of macrophages to irradiation is not fully understood. Using primary human macrophages and a monocytic cell line, we demonstrated that gamma radiation doses activate expression of various human endogenous retroviruses (HERVs). At the molecular level, we have shown that increased numbers of sense and antisense transcripts of tested HERV subgroups bind to double-stranded RNA receptors inducing the expression of type I interferons, multiple pro-inflammatory and some anti-inflammatory factors. At the phenotypic level, polarized macrophages exhibit a potent inflammatory response along with potentially tumorigenic characteristics. Our data suggest that endogenous retroviruses represent an important contributor of the macrophage-mediated inflammation in response to radiation-induced stress but may also indirectly influence tumorigenesis via biased macrophage polarization.
Collapse
Affiliation(s)
- Natallia Mikhalkevich
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Ina P. O’Carroll
- Department of Chemistry, United States Naval Academy, Annapolis, Maryland, United States of America
| | - Rok Tkavc
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Kateryna Lund
- Biomedical Instrumentation Center, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Gauthaman Sukumar
- The American Genome Center (TAGC), Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Clifton L. Dalgard
- The American Genome Center (TAGC), Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Kory R. Johnson
- Bioinformatics Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Wenxue Li
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tongguang Wang
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (AN); (SI)
| | - Sergey Iordanskiy
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- * E-mail: (AN); (SI)
| |
Collapse
|
10
|
Stroup BM, Marom R, Li X, Hsu CW, Chang CY, Truong LD, Dawson B, Grafe I, Chen Y, Jiang MM, Lanza D, Green JR, Sun Q, Barrish JP, Ani S, Christiansen AE, Seavitt JR, Dickinson ME, Kheradmand F, Heaney JD, Lee B, Burrage LC. A global Slc7a7 knockout mouse model demonstrates characteristic phenotypes of human lysinuric protein intolerance. Hum Mol Genet 2020; 29:2171-2184. [PMID: 32504080 PMCID: PMC7399531 DOI: 10.1093/hmg/ddaa107] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/30/2020] [Accepted: 05/19/2020] [Indexed: 12/18/2022] Open
Abstract
Lysinuric protein intolerance (LPI) is an inborn error of cationic amino acid (arginine, lysine, ornithine) transport caused by biallelic pathogenic variants in SLC7A7, which encodes the light subunit of the y+LAT1 transporter. Treatments for the complications of LPI, including growth failure, renal disease, pulmonary alveolar proteinosis, autoimmune disorders and osteoporosis, are limited. Given the early lethality of the only published global Slc7a7 knockout mouse model, a viable animal model to investigate global SLC7A7 deficiency is needed. Hence, we generated two mouse models with global Slc7a7 deficiency (Slc7a7em1Lbu/em1Lbu; Slc7a7Lbu/Lbu and Slc7a7em1(IMPC)Bay/em1(IMPC)Bay; Slc7a7Bay/Bay) using CRISPR/Cas9 technology by introducing a deletion of exons 3 and 4. Perinatal lethality was observed in Slc7a7Lbu/Lbu and Slc7a7Bay/Bay mice on the C57BL/6 and C57BL/6NJ inbred genetic backgrounds, respectively. We noted improved survival of Slc7a7Lbu/Lbu mice on the 129 Sv/Ev × C57BL/6 F2 background, but postnatal growth failure occurred. Consistent with human LPI, these Slc7a7Lbu/Lbu mice exhibited reduced plasma and increased urinary concentrations of the cationic amino acids. Histopathological assessment revealed loss of brush border and lipid vacuolation in the renal cortex of Slc7a7Lbu/Lbu mice, which combined with aminoaciduria suggests proximal tubular dysfunction. Micro-computed tomography of L4 vertebrae and skeletal radiographs showed delayed skeletal development and suggested decreased mineralization in Slc7a7Lbu/Lbu mice, respectively. In addition to delayed skeletal development and delayed development in the kidneys, the lungs and liver were observed based on histopathological assessment. Overall, our Slc7a7Lbu/Lbu mouse model on the F2 mixed background recapitulates multiple human LPI phenotypes and may be useful for future studies of LPI pathology.
Collapse
Affiliation(s)
- Bridget M Stroup
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ronit Marom
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children's Hospital, Houston, TX 77030, USA
| | - Xiaohui Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chih-Wei Hsu
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cheng-Yen Chang
- Department of Medicine-Pulmonary, Baylor College of Medicine, Houston, TX 77030, USA
| | - Luan D Truong
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Brian Dawson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ingo Grafe
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Center for Healthy Aging, University Clinic, Dresden D-01307, Germany
| | - Yuqing Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ming-Ming Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Denise Lanza
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jennie Rose Green
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qin Sun
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Baylor Genetics, Houston, TX 77021, USA
| | - J P Barrish
- Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Safa Ani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Audrey E Christiansen
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - John R Seavitt
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mary E Dickinson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Farrah Kheradmand
- Department of Medicine-Pulmonary, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lindsay C Burrage
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children's Hospital, Houston, TX 77030, USA
| |
Collapse
|
11
|
Nakamura T, Yamada K, Fujiwara Y, Sato Y, Harashima H. Reducing the Cytotoxicity of Lipid Nanoparticles Associated with a Fusogenic Cationic Lipid in a Natural Killer Cell Line by Introducing a Polycation-Based siRNA Core. Mol Pharm 2018; 15:2142-2150. [PMID: 29668291 DOI: 10.1021/acs.molpharmaceut.7b01166] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introducing siRNA into human immune cells by an artificial delivery system continues to be a challenging issue. We previously developed a multifunctional envelope-type nanodevice (MEND) containing the YSK12-C4, a fusogenic cationic lipid, (YSK12-MEND) and succeeded in the efficient delivery of siRNA into human immune cell lines. Significant cytotoxicity, however, was observed at siRNA doses needed for gene silencing in NK-92 cells. NK-92 cells, a unique natural killer (NK) cell line, would be applicable for use in clinical NK therapy. Thus, reducing the cytotoxicity of the YSK12-MEND in NK-92 cells would strengthen the efficacy of NK-92 cell-based therapy. The amount of the YSK12-C4 in the MEND needed to be reduced to reduce the cytotoxicity, because the cytotoxicity was directly associated with the YSK12-C4. In the present study, we decreased the total amount of lipid, including the YSK12-C4, by introducing a core formed by electrostatic interactions of siRNA with a polycation (protamine) (siRNA core), which led to a decrease in cytotoxicity in NK-92 cells. We prepared a YSK12-MEND containing an siRNA core (YSK12-MEND/core) at charge ratios (CR: YSK12-C4/siRNA) of 10, 5, 3, and 2.5 and compared the YSK12-MEND/core with that for a YSK12-MEND (CR16.9). Cell viability was increased by more than 2 times at a CR5 or less. On the other hand, the YSK12-MEND/core (CR5) maintained the same gene silencing efficiency (60%) as the YSK12-MEND. Interestingly, the cellular uptake efficiency and hemolytic activity of the YSK12-MEND/core (CR5) was reduced compared to that for the YSK12-MEND. In calculating the silencing activity per cellular uptake efficiency and hemolytic activity, the value for the YSK12-MEND/core (CR5) was more than 2 times as high as that of the YSK12-MEND. The fact indicates that after endosomal escape, the process can be enhanced by using a YSK12-MEND/core (CR5). Thus, introducing an siRNA core into lipid nanoparticles can be a potent strategy for decreasing cytotoxicity without an appreciable loss of gene silencing activity in NK-92 cells.
Collapse
Affiliation(s)
- Takashi Nakamura
- Faculty of Pharmaceutical Sciences , Hokkaido University , Kita-12, Nishi-6 , Kita-ku, Sapporo 060-0812 , Japan
| | - Koharu Yamada
- Faculty of Pharmaceutical Sciences , Hokkaido University , Kita-12, Nishi-6 , Kita-ku, Sapporo 060-0812 , Japan
| | - Yuki Fujiwara
- Faculty of Pharmaceutical Sciences , Hokkaido University , Kita-12, Nishi-6 , Kita-ku, Sapporo 060-0812 , Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences , Hokkaido University , Kita-12, Nishi-6 , Kita-ku, Sapporo 060-0812 , Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences , Hokkaido University , Kita-12, Nishi-6 , Kita-ku, Sapporo 060-0812 , Japan
| |
Collapse
|
12
|
Rotoli BM, Barilli A, Visigalli R, Ingoglia F, Milioli M, Di Lascia M, Riccardi B, Puccini P, Dall'Asta V. Downregulation of SLC7A7 Triggers an Inflammatory Phenotype in Human Macrophages and Airway Epithelial Cells. Front Immunol 2018; 9:508. [PMID: 29616026 PMCID: PMC5868322 DOI: 10.3389/fimmu.2018.00508] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/26/2018] [Indexed: 11/29/2022] Open
Abstract
Lysinuric protein intolerance (LPI) is a recessively inherited aminoaciduria caused by mutations of SLC7A7, the gene encoding y+LAT1 light chain of system y+L for cationic amino acid transport. The pathogenesis of LPI is still unknown. In this study, we have utilized a gene silencing approach in macrophages and airway epithelial cells to investigate whether complications affecting lung and immune system are directly ascribable to the lack of SLC7A7 or, rather, mediated by an abnormal accumulation of arginine in mutated cells. When SLC7A7/y+LAT1 was silenced in human THP-1 macrophages and A549 airway epithelial cells by means of short interference RNA (siRNA), a significant induction of the expression and release of the inflammatory mediators IL1β and TNFα was observed, no matter the intracellular arginine availability. This effect was mainly regulated at transcriptional level through the activation of NFκB signaling pathway. Moreover, since respiratory epithelial cells are the important sources of chemokines in response to pro-inflammatory stimuli, the effect of IL1β has been addressed on SLC7A7 silenced A549 cells. Results obtained indicated that the downregulation of SLC7A7/y+LAT1 markedly strengthened the stimulatory effect of the cytokine on CCL5/RANTES expression and release without affecting the levels of CXCL8/IL8. Consistently, also the conditioned medium of silenced THP-1 macrophages activated airway epithelial cells in terms of CCL5/RANTES expression due to the presence of elevated amount of proinflammatory cytokines. In conclusion, our results point to a novel thus far unknown function of SLC7A7/y+LAT1, that, under physiological conditions, besides transporting arginine, may act as a brake to restrain inflammation.
Collapse
Affiliation(s)
- Bianca Maria Rotoli
- Department of Medicine and Surgery (DiMeC), University of Parma, Parma, Italy
| | - Amelia Barilli
- Department of Medicine and Surgery (DiMeC), University of Parma, Parma, Italy
| | - Rossana Visigalli
- Department of Medicine and Surgery (DiMeC), University of Parma, Parma, Italy
| | - Filippo Ingoglia
- Department of Medicine and Surgery (DiMeC), University of Parma, Parma, Italy
| | - Marco Milioli
- Preclinical Pharmacokinetics, Biochemistry and Metabolism Department, Chiesi Farmaceutici, Parma, Italy
| | - Maria Di Lascia
- Preclinical Pharmacokinetics, Biochemistry and Metabolism Department, Chiesi Farmaceutici, Parma, Italy
| | - Benedetta Riccardi
- Preclinical Pharmacokinetics, Biochemistry and Metabolism Department, Chiesi Farmaceutici, Parma, Italy
| | - Paola Puccini
- Preclinical Pharmacokinetics, Biochemistry and Metabolism Department, Chiesi Farmaceutici, Parma, Italy
| | - Valeria Dall'Asta
- Department of Medicine and Surgery (DiMeC), University of Parma, Parma, Italy
| |
Collapse
|
13
|
Abstract
Traditionally cellular respiration or metabolism has been viewed as catabolic and anabolic pathways generating energy and biosynthetic precursors required for growth and general cellular maintenance. However, growing literature provides evidence of a much broader role for metabolic reactions and processes in controlling immunological effector functions. Much of this research into immunometabolism has focused on macrophages, cells that are central in pro- as well as anti-inflammatory responses—responses that in turn are a direct result of metabolic reprogramming. As we learn more about the precise role of metabolic pathways and pathway intermediates in immune function, a novel opportunity to target immunometabolism therapeutically has emerged. Here, we review the current understanding of the regulation of macrophage function through metabolic remodeling.
Collapse
Affiliation(s)
- Ciana Diskin
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Science Institute, Dublin, Ireland
| | - Eva M Pålsson-McDermott
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Science Institute, Dublin, Ireland
| |
Collapse
|
14
|
Barilli A, Gaiani F, Prandi B, Cirlini M, Ingoglia F, Visigalli R, Rotoli BM, de'Angelis N, Sforza S, de'Angelis GL, Dall'Asta V. Gluten peptides drive healthy and celiac monocytes toward an M2-like polarization. J Nutr Biochem 2017; 54:11-17. [PMID: 29216605 DOI: 10.1016/j.jnutbio.2017.10.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 10/20/2017] [Accepted: 10/26/2017] [Indexed: 01/16/2023]
Abstract
Celiac disease (CD) is an immune-mediated enteropathy triggered by ingested gluten in genetically susceptible individuals and sustained by both adaptive and innate immune responses. Recent studies in murine macrophages demonstrated that the activation of arginase (ARG) metabolic pathway by gluten peptides contributes to the modulation of intestinal permeability in vitro. Here we characterize the effects of gluten on arginine metabolism and cell polarization in human monocytes from both healthy and CD subjects; both a simplified enzymatic digestion of gliadin and a physiological digestion of whole wheat have been tested. Results indicate that gluten digests induce the onset of an M2-like phenotype in activated macrophages; more precisely, both isoforms of arginase, ARG1 and ARG2, are induced likely due to the inhibition of mTOR and the consequent induction of C/EBPβ transcription factor. These effects are independent from the origin of gluten as well as from the digestive protocol employed; moreover, no statistical difference can be evidenced between healthy and CD patients, excluding a diverse predisposition of CD monocytes to gluten-triggered polarization with respect to healthy immune cells. Overall, the present findings sustain a role for arginase pathway in the immune response elicited by human monocytes toward ingested gluten that, hence, deserves particular attention when addressing the pathogenesis of CD.
Collapse
Affiliation(s)
- Amelia Barilli
- Unit of General Pathology, Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Federica Gaiani
- Unit of Gastroenterology and Digestive Endoscopy of Parma, University Hospital of Parma, via Gramsci 14, 43126 Parma, Italy
| | - Barbara Prandi
- Department of Food Science, University of Parma, Parco Area delle Scienze 17A, 43124 Parma, Italy
| | - Martina Cirlini
- Department of Food Science, University of Parma, Parco Area delle Scienze 17A, 43124 Parma, Italy
| | - Filippo Ingoglia
- Unit of General Pathology, Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Rossana Visigalli
- Unit of General Pathology, Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Bianca Maria Rotoli
- Unit of General Pathology, Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Nicola de'Angelis
- Department of HPB Surgery and Liver Transplantation, Henri-Mondor Hospital, Université Paris Est-UPEC, Créteil, France
| | - Stefano Sforza
- Department of Food Science, University of Parma, Parco Area delle Scienze 17A, 43124 Parma, Italy
| | - Gian Luigi de'Angelis
- Unit of Gastroenterology and Digestive Endoscopy of Parma, University Hospital of Parma, via Gramsci 14, 43126 Parma, Italy
| | - Valeria Dall'Asta
- Unit of General Pathology, Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Via Volturno 39, 43125 Parma, Italy.
| |
Collapse
|
15
|
Saraiva JP, Oswald M, Biering A, Röll D, Assmann C, Klassert T, Blaess M, Czakai K, Claus R, Löffler J, Slevogt H, König R. Fungal biomarker discovery by integration of classifiers. BMC Genomics 2017; 18:601. [PMID: 28797245 PMCID: PMC5553868 DOI: 10.1186/s12864-017-4006-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 08/02/2017] [Indexed: 02/01/2023] Open
Abstract
Background The human immune system is responsible for protecting the host from infection. However, in immunocompromised individuals the risk of infection increases substantially with possible drastic consequences. In extreme, systemic infection can lead to sepsis which is responsible for innumerous deaths worldwide. Amongst its causes are infections by bacteria and fungi. To increase survival, it is mandatory to identify the type of infection rapidly. Discriminating between fungal and bacterial pathogens is key to determine if antifungals or antibiotics should be administered, respectively. For this, in situ experiments have been performed to determine regulation mechanisms of the human immune system to identify biomarkers. However, these studies led to heterogeneous results either due different laboratory settings, pathogen strains, cell types and tissues, as well as the time of sample extraction, to name a few. Methods To generate a gene signature capable of discriminating between fungal and bacterial infected samples, we employed Mixed Integer Linear Programming (MILP) based classifiers on several datasets comprised of the above mentioned pathogens. Results When combining the classifiers by a joint optimization we could increase the consistency of the biomarker gene list independently of the experimental setup. An increase in pairwise overlap (the number of genes that overlap in each cross-validation) of 43% was obtained by this approach when compared to that of single classifiers. The refined gene list was composed of 19 genes and ranked according to consistency in expression (up- or down-regulated) and most of them were linked either directly or indirectly to the ERK-MAPK signalling pathway, which has been shown to play a key role in the immune response to infection. Testing of the identified 12 genes on an unseen dataset yielded an average accuracy of 83%. Conclusions In conclusion, our method allowed the combination of independent classifiers and increased consistency and reliability of the generated gene signatures. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-4006-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- João Pedro Saraiva
- Network Modelling, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Beutenbergstraße 11a, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Marcus Oswald
- Network Modelling, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Beutenbergstraße 11a, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Antje Biering
- Network Modelling, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Beutenbergstraße 11a, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Daniela Röll
- Network Modelling, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Beutenbergstraße 11a, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Cora Assmann
- Septomics Research Centre, Jena University Hospital, Jena, Germany
| | - Tilman Klassert
- Septomics Research Centre, Jena University Hospital, Jena, Germany
| | - Markus Blaess
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | | | - Ralf Claus
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | | | - Hortense Slevogt
- Septomics Research Centre, Jena University Hospital, Jena, Germany
| | - Rainer König
- Network Modelling, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Beutenbergstraße 11a, Jena, Germany. .,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.
| |
Collapse
|
16
|
Nakamura T, Kuroi M, Fujiwara Y, Warashina S, Sato Y, Harashima H. Small-sized, stable lipid nanoparticle for the efficient delivery of siRNA to human immune cell lines. Sci Rep 2016; 6:37849. [PMID: 27892533 PMCID: PMC5124971 DOI: 10.1038/srep37849] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 10/31/2016] [Indexed: 02/05/2023] Open
Abstract
Gene silencing by small interfering RNA (siRNA) is useful for analyzing the functions of human immune cells. However, the transfection of siRNA to human immune cells is difficult. Here, we used a multifunctional envelope-type nanodevice (MEND) containing YSK12-C4 (YSK12-MEND) to efficiently introduce siRNA to human immune cell lines, Jurkat, THP-1, KG-1 and NK92. The YSK12-MEND was transfected to human immune cell lines at a siRNA dose range of 1–30 nM, resulting that maximum gene silencing efficiencies at the mRNA level in Jurkat, THP-1, KG-1 and NK92 were 96%, 96%, 91% and 75%, respectively. The corresponding values for Lipofectamine RNAiMAX (RNAiMAX) were 37%, 56%, 43% and 19%, respectively. The process associated with cellular uptake played a role in effective gene silencing effect of the YSK12-MEND. The small size and high non-aggregability of the YSK12-MEND were advantageous for the cellular internalization of siRNA to immune cell lines. In the case of RNAiMAX, a drastic increase in particles size was observed in the medium used, which inhibited cellular uptake. The YSK12-MEND reported in herein appears to be appropriate for delivering siRNA to human immune cells, and the small particle size and non-aggregability are essential properties.
Collapse
Affiliation(s)
- Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Moeka Kuroi
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Yuki Fujiwara
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Shota Warashina
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| |
Collapse
|
17
|
Inhaled Sargramostim Induces Resolution of Pulmonary Alveolar Proteinosis in Lysinuric Protein Intolerance. JIMD Rep 2016; 34:97-104. [PMID: 27783330 DOI: 10.1007/8904_2016_15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/29/2016] [Accepted: 09/14/2016] [Indexed: 12/13/2022] Open
Abstract
Pulmonary alveolar proteinosis (PAP) is a potentially fatal complication of lysinuric protein intolerance (LPI), an inherited disorder of cationic amino acid transport. The patients often present with mild respiratory symptoms, which may rapidly progress to acute respiratory failure responding poorly to conventional treatment with steroids and bronchoalveolar lavations (BALs). The pathogenesis of PAP in LPI is still largely unclear. In previous studies, we have shown disturbances in the function and activity of alveolar macrophages of these patients, suggesting that increasing the activity and the number of macrophages by recombinant human GM-CSF (rhuGM-CSF) might be beneficial in this patient group.Two LPI patients with complicated PAP were treated with experimental inhaled rhuGM-CSF (sargramostim) after poor response to maximal conventional therapy. BAL fluid and cell samples from one patient were studied with light microscopy and transmission electron microscopy.Excellent response to therapy was observed in patient 1 with no compliance problems or side effects. Macrophages with myelin figure-like structures were seen in her BAL sample. Slight improvement of the pulmonary function was evident also in patient 2, but the role of sargramostim could not be properly evaluated due to the complicated clinical situation.In conclusion, inhaled rhuGM-CSF might be of benefit in patients with LPI-associated PAP.
Collapse
|
18
|
IRAK regulates macrophage foam cell formation by modulating genes involved in cholesterol uptake and efflux. Bioessays 2016; 38:591-604. [DOI: 10.1002/bies.201600085] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
19
|
Labranche N, Khattabi CE, Berkenboom G, Pochet S. Effects of diesel exhaust particles on macrophage polarization. Hum Exp Toxicol 2016; 36:412-420. [PMID: 27224998 DOI: 10.1177/0960327116651123] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Exposure to diesel exhaust particles (DEP) has long been associated with increased cardiovascular morbidity and mortality. The development of DEP toxicity seems to be linked to inflammation in which macrophages play a critical role. Macrophages can be polarized into proinflammatory M1 or anti-inflammatory M2 macrophages. The aim of this study was to identify the role of inflammation in DEP-induced toxicity by assessing the effects of DEP on macrophage polarization. METHODS Monocyte-derived macrophages (Mϕ) were stimulated with interferon γ and lipopolysaccharide or interleukin (IL)-4 to obtain M1 and M2 subtypes, respectively. To test the polarization capacity of DEP, Mϕ cells were exposed to DEP and compared to Mϕ, M1, and M2. We also studied the effects of DEP on already-polarized M1 or M2. The M1 markers assessed were tumor necrosis factor α (TNF-α) and IL-1β, while the M2 markers were the mannose receptor C type 1 (MRC-1) and transglutaminase 2 (TGM2). RESULTS Western blots revealed a 31 kDa band corresponding to pro-IL-1β, but only in M1-polarized macrophages. In M1, we also observed an upregulation of TNF-α messenger RNA (mRNA) expression. MRC-1 and TGM2 mRNA expression were only significantly enhanced in M2. DEP had no effect on any of the M1/M2 markers assessed. Moreover, DEP were not able to modify the phenotype of already-polarized M1 or M2. CONCLUSION Mϕ incubation with DEP did not have any effect on macrophage polarization, at least on the markers assessed in this study, namely, TNF-α/IL-1β for M1, and MRC-1/TGM2 for M2. Hence, these data argue against an important role of inflammation in DEP-induced vascular toxicity.
Collapse
Affiliation(s)
- N Labranche
- 1 Laboratory of Physiology and Pharmacology, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - C El Khattabi
- 1 Laboratory of Physiology and Pharmacology, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - G Berkenboom
- 2 Department of Cardiology, Erasme Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - S Pochet
- 1 Laboratory of Physiology and Pharmacology, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
20
|
Hu Y, He MY, Zhu LF, Yang CC, Zhou ML, Wang Q, Zhang W, Zheng YY, Wang DM, Xu ZQ, Wu YN, Liu LK. Tumor-associated macrophages correlate with the clinicopathological features and poor outcomes via inducing epithelial to mesenchymal transition in oral squamous cell carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:12. [PMID: 26769084 PMCID: PMC4714460 DOI: 10.1186/s13046-015-0281-z] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/30/2015] [Indexed: 01/05/2023]
Abstract
Background Both tumor-associated macrophages (TAMs) and the epithelial to mesenchymal transition (EMT) of cancer cells play key roles in promoting tumor progression. However, whether TAMs could induce EMT in the progression of oral squamous cell carcinoma (OSCC) remains undefined. Results Here we detected the expression of macrophages markers CD68 and CD163, epithelial marker E-cadherin and mesenchymal marker vimentin in 127 OSCC patients by using semi-quantitative immunohistochemistry. CD68 and CD163 expression was not confined to the infiltrating TAMs, but also detected in cancer cells. The high number of CD68-positive macrophages was correlated with poor overall survival. Meanwhile, the expression of CD163 both in macrophages and in cancer cells was associated with poor overall survival and had a significant prognostic impact in OSCC. Importantly, the expression of CD163 in cancer cells had a significant relationship with E-cadherin and vimentin. Furthermore, the incubation of TAMs conditioned medium resulted in a fibroblast-like appearance of cancer cells (HN4, HN6 and SCC9) together with the decreased/increased expression of E-cadherin/ vimentin, which were correlated with the enhanced ability of migration and invasion. Conclusions Our results indicate that TAMs could promote the EMT of cancer cells, thereby leading to the progression of oral cancer.
Collapse
Affiliation(s)
- Yong Hu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Postal#210029 136# Hanzhong Road, Nanjing, Jiangsu, the People's Republic of China. .,Department of Stomatology, Suzhou Kowloon Hospital Shanghai Jiaotong University Medical School, Suzhou, People's Republic of China.
| | - Meng-Ying He
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Postal#210029 136# Hanzhong Road, Nanjing, Jiangsu, the People's Republic of China.
| | - Li-Fang Zhu
- Department of Stomatology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.
| | - Cong-Chong Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Postal#210029 136# Hanzhong Road, Nanjing, Jiangsu, the People's Republic of China.
| | - Mei-Ling Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Postal#210029 136# Hanzhong Road, Nanjing, Jiangsu, the People's Republic of China.
| | - Qiong Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Postal#210029 136# Hanzhong Road, Nanjing, Jiangsu, the People's Republic of China.
| | - Wei Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Postal#210029 136# Hanzhong Road, Nanjing, Jiangsu, the People's Republic of China.
| | - Yang-Yu Zheng
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Postal#210029 136# Hanzhong Road, Nanjing, Jiangsu, the People's Republic of China.
| | - Dong-Miao Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Postal#210029 136# Hanzhong Road, Nanjing, Jiangsu, the People's Republic of China.
| | - Zeng-Qi Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Postal#210029 136# Hanzhong Road, Nanjing, Jiangsu, the People's Republic of China.
| | - Yu-Nong Wu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Postal#210029 136# Hanzhong Road, Nanjing, Jiangsu, the People's Republic of China.
| | - Lai-Kui Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Basic Science of Stomatology, Affiliated Hospital of Stomatology, Nanjing Medical University, Postal#210029 136# Hanzhong Road, Nanjing, Jiangsu, the People's Republic of China.
| |
Collapse
|
21
|
Costa GNO, Dudbridge F, Fiaccone RL, da Silva TM, Conceição JS, Strina A, Figueiredo CA, Magalhães WCS, Rodrigues MR, Gouveia MH, Kehdy FSG, Horimoto ARVR, Horta B, Burchard EG, Pino-Yanes M, Del Rio Navarro B, Romieu I, Hancock DB, London S, Lima-Costa MF, Pereira AC, Tarazona E, Rodrigues LC, Barreto ML. A genome-wide association study of asthma symptoms in Latin American children. BMC Genet 2015; 16:141. [PMID: 26635092 PMCID: PMC4669662 DOI: 10.1186/s12863-015-0296-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/17/2015] [Indexed: 01/14/2023] Open
Abstract
Background Asthma is a chronic disease of the airways and, despite the advances in the knowledge of associated genetic regions in recent years, their mechanisms have yet to be explored. Several genome-wide association studies have been carried out in recent years, but none of these have involved Latin American populations with a high level of miscegenation, as is seen in the Brazilian population. Methods 1246 children were recruited from a longitudinal cohort study in Salvador, Brazil. Asthma symptoms were identified in accordance with an International Study of Asthma and Allergies in Childhood (ISAAC) questionnaire. Following quality control, 1 877 526 autosomal SNPs were tested for association with childhood asthma symptoms by logistic regression using an additive genetic model. We complemented the analysis with an estimate of the phenotypic variance explained by common genetic variants. Replications were investigated in independent Mexican and US Latino samples. Results Two chromosomal regions reached genome-wide significance level for childhood asthma symptoms: the 14q11 region flanking the DAD1 and OXA1L genes (rs1999071, MAF 0.32, OR 1.78, 95 % CI 1.45–2.18, p-value 2.83 × 10−8) and 15q22 region flanking the FOXB1 gene (rs10519031, MAF 0.04, OR 3.0, 95 % CI 2.02–4.49, p-value 6.68 × 10−8 and rs8029377, MAF 0.03, OR 2.49, 95 % CI 1.76–3.53, p-value 2.45 × 10−7). eQTL analysis suggests that rs1999071 regulates the expression of OXA1L gene. However, the original findings were not replicated in the Mexican or US Latino samples. Conclusions We conclude that the 14q11 and 15q22 regions may be associated with asthma symptoms in childhood. Electronic supplementary material The online version of this article (doi:10.1186/s12863-015-0296-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gustavo N O Costa
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil.
| | - Frank Dudbridge
- Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK.
| | | | - Thiago M da Silva
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil.
| | | | - Agostino Strina
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil.
| | - Camila A Figueiredo
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.
| | - Wagner C S Magalhães
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Maira R Rodrigues
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Mateus H Gouveia
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Fernanda S G Kehdy
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | | | - Bernardo Horta
- Programa de Pós Graduação em Epidemiologia, Universidade Federal de Pelotas, Pelotas, Brazil.
| | | | - Maria Pino-Yanes
- Department of Medicine, University of California, San Francisco, USA.
| | - Blanca Del Rio Navarro
- Department of Health and Human Services, Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA.
| | | | - Dana B Hancock
- Behavioral and Urban Health Program, Research Triangle Institute (RTI) International, Research Triangle Park, North Carolina, USA.
| | - Stephanie London
- Department of Health and Human Services, Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA.
| | | | - Alexandre C Pereira
- Instituto de Pesquisas Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil.
| | - Eduardo Tarazona
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Laura C Rodrigues
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK.
| | - Mauricio L Barreto
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil. .,Centro de Pesquisa Gonçalo Muniz, Fundação Osvaldo Cruz, Salvador, Brazil.
| |
Collapse
|
22
|
Ingoglia F, Visigalli R, Rotoli BM, Barilli A, Riccardi B, Puccini P, Dall'Asta V. Functional activity of L-carnitine transporters in human airway epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:210-9. [PMID: 26607009 DOI: 10.1016/j.bbamem.2015.11.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/21/2015] [Accepted: 11/18/2015] [Indexed: 12/30/2022]
Abstract
Carnitine plays a physiologically important role in the β-oxidation of fatty acids, facilitating the transport of long-chain fatty acids across the inner mitochondrial membrane. Distribution of carnitine within the body tissues is mainly performed by novel organic cation transporter (OCTN) family, including the isoforms OCTN1 (SLC22A4) and OCTN2 (SLC22A5) expressed in human. We performed here a characterization of carnitine transport in human airway epithelial cells A549, Calu-3, NCl-H441, and BEAS-2B, by means of an integrated approach combining data of mRNA/protein expression with the kinetic and inhibition analyses of L-[(3)H]carnitine transport. Carnitine uptake was strictly Na(+)-dependent in all cell models. In A549 and BEAS-2B cells, carnitine uptake was mediated by one high-affinity component (Km<2 μM) identifiable with OCTN2. In both these cell models, indeed, carnitine uptake was maximally inhibited by betaine and strongly reduced by SLC22A5/OCTN2 silencing. Conversely, Calu-3 and NCl-H441 exhibited both a high (Km~20 μM) and a low affinity (Km>1 mM) transport component. While the high affinity component is identifiable with OCTN2, the low affinity uptake is mediated by ATB(0,+), a Na(+), and Cl(-)-coupled transport system for neutral and cationic amino acids, as demonstrated by the inhibition by leucine and arginine, as well as by SLC6A14/ATB(0,+) silencing. The presence of this transporter leads to a massive accumulation of carnitine inside the cells and may be of peculiar relevance in pathologic conditions of carnitine deficiency, such as those associated to OCTN2 defects.
Collapse
Affiliation(s)
- Filippo Ingoglia
- Dept. of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Rossana Visigalli
- Dept. of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Bianca Maria Rotoli
- Dept. of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Amelia Barilli
- Dept. of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Benedetta Riccardi
- Preclinical Pharmacokinetics, Biochemistry & Metabolism Dept., Chiesi Farmaceutici, Largo F. Belloli 11/A, 43122 Parma, Italy
| | - Paola Puccini
- Preclinical Pharmacokinetics, Biochemistry & Metabolism Dept., Chiesi Farmaceutici, Largo F. Belloli 11/A, 43122 Parma, Italy
| | - Valeria Dall'Asta
- Dept. of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Via Volturno 39, 43125 Parma, Italy.
| |
Collapse
|
23
|
Kurko J, Vähä-Mäkilä M, Tringham M, Tanner L, Paavanen-Huhtala S, Saarinen M, Näntö-Salonen K, Simell O, Niinikoski H, Mykkänen J. Dysfunction in macrophage toll-like receptor signaling caused by an inborn error of cationic amino acid transport. Mol Immunol 2015. [PMID: 26210182 DOI: 10.1016/j.molimm.2015.07.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Amino acids, especially arginine, are vital for the well-being and activity of immune cells, and disruption of amino acid balance may weaken immunity and predispose to infectious and autoimmune diseases. We present here a model of an inborn aminoaciduria, lysinuric protein intolerance (LPI), in which a single mutation in y(+)LAT1 cationic amino acid transporter gene SLC7A7 leads to a multisystem disease characterized by immunological complications, life-threatening pulmonary alveolar proteinosis and nephropathy. Macrophages are suggested to play a central role in LPI in the development of these severe secondary symptoms. We thus studied the effect of the Finnish y(+)LAT1 mutation on monocyte-derived macrophages where toll-like receptors (TLRs) act as the key molecules in innate immune response against external pathogens. The function of LPI patient and control macrophage TLR signaling was examined by stimulating the TLR2/1, TLR4 and TLR9 pathways with their associated pathogen-associated molecular patterns. Downregulation in expression of TLR9, IRF7, IRF3 and IFNB1 and in secretion of IFN-α was detected, suggesting an impaired response to TLR9 stimulation. In addition, secretion of TNF-α, IL-12 and IL-1RA by TLR2/1 stimulation and IL-12 and IL-1RA by TLR4 stimulation was increased in the LPI patients. LPI macrophages secreted significantly less nitric oxide than control macrophages, whereas plasma concentrations of inflammatory chemokines CXCL8, CXCL9 and CXCL10 were elevated in the LPI patients. In conclusion, our results strengthen the relevance of macrophages in the pathogenesis of LPI and, furthermore, suggest that cationic amino acid transport plays an important role in the regulation of innate immune responses.
Collapse
Affiliation(s)
- Johanna Kurko
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland.
| | - Mari Vähä-Mäkilä
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Maaria Tringham
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland.
| | - Laura Tanner
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland; Department of Clinical Genetics, Turku University Hospital, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Sari Paavanen-Huhtala
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Maiju Saarinen
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland; Department of Public Health, University of Turku, Lemminkäisenkatu 1, 20014 Turku, Finland.
| | - Kirsti Näntö-Salonen
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Olli Simell
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Harri Niinikoski
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Juha Mykkänen
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland; Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland.
| |
Collapse
|
24
|
Pekarova M, Lojek A. The crucial role of l-arginine in macrophage activation: What you need to know about it. Life Sci 2015; 137:44-8. [PMID: 26188591 DOI: 10.1016/j.lfs.2015.07.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 07/07/2015] [Accepted: 07/11/2015] [Indexed: 11/19/2022]
Abstract
Nowadays, it is well recognized that amino acids are powerful molecules responsible for regulatory control over fundamental cellular processes. However, our understanding of the signaling cascades involved in amino acid sensing in organisms, as well as signal initiation, is largely limited. This is also the case of semi-essential amino acid l-arginine, which has multiple metabolic fates, and it is considered as one of the most versatile amino acids. Recently, some new and important facts have been published considering the role of l-arginine in the regulation of inflammatory processes in several human and mouse models, mediated also via the regulation of macrophage activation. Therefore, this mini review focuses on the actual summarization of information about (i) l-arginine bioavailability in organism, (ii) l-arginine-dependent regulation of nitric oxide synthase expression and nitric oxide production, and importantly (iii) its role in the activation of intracellular signaling pathways and G-protein-coupled receptors in macrophages.
Collapse
Affiliation(s)
- Michaela Pekarova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65 Brno, Czech Republic.
| | - Antonin Lojek
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65 Brno, Czech Republic
| |
Collapse
|
25
|
Barilli A, Rotoli BM, Visigalli R, Ingoglia F, Cirlini M, Prandi B, Dall'Asta V. Gliadin-mediated production of polyamines by RAW264.7 macrophages modulates intestinal epithelial permeability in vitro. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1779-86. [PMID: 26047680 DOI: 10.1016/j.bbadis.2015.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 04/28/2015] [Accepted: 06/01/2015] [Indexed: 01/22/2023]
Abstract
Celiac disease (CD) is an immune-mediated enteropathy sustained by dietary gluten in susceptible individuals, and characterized by a complex interplay between adaptive and innate responses against gluten peptides (PTG). In a recent contribution we have demonstrated that the treatment with PTG induces the expression and activity of arginase in both murine macrophages and human monocytes from healthy subjects, thus suggesting a role for arginine and its metabolites in gluten-triggered response of these cells. Here we further explore this field, by addressing the effects of PTG on polyamine synthesis and release in murine RAW264.7 macrophages, and how they affect epithelial permeability of Caco-2 monolayers. Results obtained show a massive production and release of putrescine by macrophages upon incubation with gluten peptides; this, in turn, causes a decrease in TEER in epithelial cells, indicating that PTG-driven secretion of polyamines by macrophages has a role in the modulation of intestinal permeability in vitro. At a molecular level, putrescine production appears referable to the activation of C/EBPβ transcription factor, which is known to be responsible for arginase induction in activated macrophages and is a crucial mediator of inflammation. Whether these pathways are stimulated also in vivo deserves to be further investigated, as well as their role in gluten-driven cellular and intestinal defects typical of CD patients.
Collapse
Affiliation(s)
- Amelia Barilli
- Dipartimento di Scienze Biomediche, Biotecnologiche e Traslazionali, SBiBiT, Università degli Studi di Parma, Via Volturno 39, 43125 Parma, Italy
| | - Bianca Maria Rotoli
- Dipartimento di Scienze Biomediche, Biotecnologiche e Traslazionali, SBiBiT, Università degli Studi di Parma, Via Volturno 39, 43125 Parma, Italy
| | - Rossana Visigalli
- Dipartimento di Scienze Biomediche, Biotecnologiche e Traslazionali, SBiBiT, Università degli Studi di Parma, Via Volturno 39, 43125 Parma, Italy
| | - Filippo Ingoglia
- Dipartimento di Scienze Biomediche, Biotecnologiche e Traslazionali, SBiBiT, Università degli Studi di Parma, Via Volturno 39, 43125 Parma, Italy
| | - Martina Cirlini
- Dipartimento di Scienze degli Alimenti, Università degli Studi di Parma, Parco Area delle Scienze 49/A, 43124 Parma, Italy
| | - Barbara Prandi
- Dipartimento di Scienze degli Alimenti, Università degli Studi di Parma, Parco Area delle Scienze 49/A, 43124 Parma, Italy
| | - Valeria Dall'Asta
- Dipartimento di Scienze Biomediche, Biotecnologiche e Traslazionali, SBiBiT, Università degli Studi di Parma, Via Volturno 39, 43125 Parma, Italy.
| |
Collapse
|
26
|
Rossi F, Casano AM, Henke K, Richter K, Peri F. The SLC7A7 Transporter Identifies Microglial Precursors prior to Entry into the Brain. Cell Rep 2015; 11:1008-17. [PMID: 25959825 DOI: 10.1016/j.celrep.2015.04.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/10/2015] [Accepted: 04/13/2015] [Indexed: 01/13/2023] Open
Abstract
During development, macrophages invade organs to establish phenotypically and transcriptionally distinct tissue-resident populations. How they invade and colonize these organs is unclear. In particular, it remains to be established whether they arise from naive equivalents that colonize organs randomly or whether there are committed macrophages that follow pre-determined migration paths. Here, by using a combination of genetics and imaging approaches in the zebrafish embryo, we have addressed how macrophages colonize the brain to become microglia. Identification and cloning of a mutant that lacks microglia has shown that Slc7a7, a Leucine/Arginine transporter, defines a restricted macrophage sub-lineage and is necessary for brain colonization. By taking a photoconversion approach, we show that these macrophages give rise to microglia. This study provides direct experimental evidence for the existence of sub-lineages among embryonic macrophages.
Collapse
Affiliation(s)
- Federico Rossi
- Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidbelberg, Germany
| | - Alessandra Maria Casano
- Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidbelberg, Germany
| | - Katrin Henke
- Department of Genetics, Max-Planck Institute (MPI) for Developmental Biology, Spemannstraße 35-39, 72076 Tübingen, Germany
| | - Kerstin Richter
- Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidbelberg, Germany
| | - Francesca Peri
- Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidbelberg, Germany.
| |
Collapse
|
27
|
Ingoglia F, Visigalli R, Rotoli BM, Barilli A, Riccardi B, Puccini P, Dall'Asta V. Functional characterization of the organic cation transporters (OCTs) in human airway pulmonary epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:1563-72. [PMID: 25883089 DOI: 10.1016/j.bbamem.2015.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 03/25/2015] [Accepted: 04/02/2015] [Indexed: 12/28/2022]
Abstract
Organic cation transporters (OCT1-3) mediate the transport of organic cations including inhaled drugs across the cell membrane, although their role in lung epithelium hasn't been well understood yet. We address here the expression and functional activity of OCT1-3 in human airway epithelial cells A549, Calu-3 and NCl-H441. Kinetic and inhibition analyses, employing [(3)H]1-methyl-4-phenylpyridinium (MPP+) as substrate, and the compounds quinidine, prostaglandine E2 (PGE2) and corticosterone as preferential inhibitors of OCT1, OCT2, and OCT3, respectively, have been performed. A549 cells present a robust MPP+ uptake mediated by one high-affinity component (Km~50μM) which is identifiable with OCT3. Corticosterone, indeed, completely inhibits MPP+ transport, while quinidine and PGE2 are inactive and SLC22A3/OCT3 silencing with siRNA markedly lowers MPP+ uptake. Conversely, Calu-3 exhibits both a high (Km<20μM) and a low affinity (Km>0.6mM) transport components, referable to OCT3 and OCT1, respectively, as demonstrated by the inhibition analysis performed at proper substrate concentrations and confirmed by the use of specific siRNA. These transporters are active also when cells are grown under air-liquid interface (ALI) conditions. Only a very modest saturable MPP+ uptake is measurable in NCl-H441 cells and the inhibitory effect of quinidine points to OCT1 as the subtype functionally involved in this model. Finally, the characterization of MPP+ transport in human bronchial BEAS-2B cells suggests that OCT1 and OCT3 are operative. These findings could help to identify in vitro models to be employed for studies concerning the specific involvement of each transporter in drug transportation.
Collapse
Affiliation(s)
- Filippo Ingoglia
- Dept. of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Rossana Visigalli
- Dept. of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Bianca Maria Rotoli
- Dept. of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Amelia Barilli
- Dept. of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Benedetta Riccardi
- Preclinical Pharmacokinetics, Biochemistry & Metabolism Dept., Chiesi Farmaceutici, Largo F. Belloli 11/A, 43122 Parma, Italy
| | - Paola Puccini
- Preclinical Pharmacokinetics, Biochemistry & Metabolism Dept., Chiesi Farmaceutici, Largo F. Belloli 11/A, 43122 Parma, Italy
| | - Valeria Dall'Asta
- Dept. of Biomedical, Biotechnological and Translational Sciences (SBiBiT), University of Parma, Via Volturno 39, 43125 Parma, Italy.
| |
Collapse
|
28
|
Mussai F, Egan S, Higginbotham-Jones J, Perry T, Beggs A, Odintsova E, Loke J, Pratt G, U KP, Lo A, Ng M, Kearns P, Cheng P, De Santo C. Arginine dependence of acute myeloid leukemia blast proliferation: a novel therapeutic target. Blood 2015; 125:2386-96. [PMID: 25710880 PMCID: PMC4416943 DOI: 10.1182/blood-2014-09-600643] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 02/11/2015] [Indexed: 12/14/2022] Open
Abstract
Acute myeloid leukemia (AML) is one of the most common acute leukemias in adults and children, yet significant numbers of patients relapse and die of disease. In this study, we identify the dependence of AML blasts on arginine for proliferation. We show that AML blasts constitutively express the arginine transporters CAT-1 and CAT-2B, and that the majority of newly diagnosed patients' blasts have deficiencies in the arginine-recycling pathway enzymes argininosuccinate synthase and ornithine transcarbamylase, making them arginine auxotrophic. BCT-100, a pegylated human recombinant arginase, leads to a rapid depletion in extracellular and intracellular arginine concentrations, resulting in arrest of AML blast proliferation and a reduction in AML engraftment in vivo. BCT-100 as a single agent causes significant death of AML blasts from adults and children, and acts synergistically in combination with cytarabine. Using RNA sequencing, 20 further candidate genes which correlated with resistance have been identified. Thus, AML blasts are dependent on arginine for survival and proliferation, as well as depletion of arginine with BCT-100 of clinical value in the treatment of AML.
Collapse
MESH Headings
- Adolescent
- Aged
- Animals
- Antimetabolites, Antineoplastic/therapeutic use
- Arginase/therapeutic use
- Arginine/metabolism
- Child
- Child, Preschool
- Cytarabine/therapeutic use
- Enzyme Therapy
- Female
- Humans
- Infant
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice, SCID
- Middle Aged
- Recombinant Proteins/therapeutic use
- Tumor Cells, Cultured
- Young Adult
Collapse
Affiliation(s)
- Francis Mussai
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Sharon Egan
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | | | - Tracey Perry
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Andrew Beggs
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Elena Odintsova
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Justin Loke
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Guy Pratt
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Kin Pong U
- Bio-cancer Treatment International Ltd, Hong Kong Science Park, Shatin, New Territories, Hong Kong; and
| | - Anthony Lo
- Department of Anatomic Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Margaret Ng
- Department of Anatomic Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Pamela Kearns
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Cheng
- Bio-cancer Treatment International Ltd, Hong Kong Science Park, Shatin, New Territories, Hong Kong; and
| | - Carmela De Santo
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
29
|
Gliadin activates arginase pathway in RAW264.7 cells and in human monocytes. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1364-71. [PMID: 24793417 DOI: 10.1016/j.bbadis.2014.04.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 04/10/2014] [Accepted: 04/25/2014] [Indexed: 11/21/2022]
Abstract
Celiac disease (CD) is an autoimmune enteropathy triggered in susceptible individuals by the ingestion of gliadin-containing grains. Recent studies have demonstrated that macrophages play a key role in the pathogenesis of CD through the release of inflammatory mediators such as cytokines and nitric oxide (NO). Since arginine is the obliged substrate of iNOS (inducible nitric oxide synthase), the enzyme that produces large amount of NO, the aim of this work is to investigate arginine metabolic pathways in RAW264.7 murine macrophages after treatment with PT-gliadin (PTG) in the absence and in the presence of IFNγ. Our results demonstrate that, besides strengthening the IFNγ-dependent activation of iNOS, gliadin is also an inducer of arginase, the enzyme that transforms arginine into ornithine and urea. Gliadin treatment increases, indeed, the expression and the activity of arginase, leading to the production of polyamines through the subsequent induction of ornithine decarboxylase. This effect is strengthened by IFNγ. The activation of these pathways takes advantage of the increased availability of arginine due to a decreased system y(+)l-mediated efflux, likely ascribable to a reduced expression of Slc7a6 transporter. A significant induction of arginase expression is also observed in human monocytes from healthy subject upon treatment with gliadin, thus demonstrating that gluten components trigger changes in arginine metabolism in monocyte/macrophage cells.
Collapse
|
30
|
Barilli A, Atzeri C, Bassanetti I, Ingoglia F, Dall'Asta V, Bussolati O, Maffini M, Mucchino C, Marchiò L. Oxidative stress induced by copper and iron complexes with 8-hydroxyquinoline derivatives causes paraptotic death of HeLa cancer cells. Mol Pharm 2014; 11:1151-63. [PMID: 24592930 DOI: 10.1021/mp400592n] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Here, we report the antiproliferative/cytotoxic properties of 8-hydroxyquinoline (8-HQ) derivatives on HeLa cells in the presence of transition metal ions (Cu(2+), Fe(3+), Co(2+), Ni(2+)). Two series of ligands were tested, the arylvinylquinolinic L1-L8 and the arylethylenequinolinic L9-L16, which can all interact with metal ions by virtue of the N,O donor set of 8-HQ; however, only L9-L16 are flexible enough to bind the metal in a multidentate fashion, thus exploiting the additional donor functions. L1-L16 were tested for their cytotoxicity on HeLa cancer cells, both in the absence and in the presence of copper. Among them, the symmetric L14 exhibits the highest differential activity between the ligand alone (IC50 = 23.7 μM) and its copper complex (IC50 = 1.8 μM). This latter, besides causing a significant reduction of cell viability, is associated with a considerable accumulation of the metal inside the cells. Metal accumulation is also observed when the cells are incubated with L14 complexed with other late transition metal ions (Fe(3+), Co(2+), Ni(2+)), although the biological response of HeLa cells is different. In fact, while Ni/L14 and Co/L14 exert a cytostatic effect, both Cu/L14 and Fe/L14 trigger a caspase-independent paraptotic process, which results from the induction of a severe oxidative stress and the unfolded protein response.
Collapse
Affiliation(s)
- Amelia Barilli
- Dipartimento di Chimica, Università degli Studi di Parma , Viale delle Scienze 17/A, 43123 Parma, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Yang J, Tan Q, Zhu W, Chen C, Liang X, Pan L. Cloning and molecular characterization of cationic amino acid transporter y⁺LAT1 in grass carp (Ctenopharyngodon idellus). FISH PHYSIOLOGY AND BIOCHEMISTRY 2014; 40:93-104. [PMID: 23817987 DOI: 10.1007/s10695-013-9827-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 06/18/2013] [Indexed: 06/02/2023]
Abstract
The solute carrier family 7A, member 7 gene encodes the light chain- y⁺L amino acid transporter-1 (y⁺LAT1) of the heterodimeric carrier responsible for cationic amino acid (CAA) transport across the basolateral membranes of epithelial cells in intestine and kidney. Rising attention has been given to y⁺LAT1 involved in CAA metabolic pathways and growth control. The molecular characterization and function analysis of y⁺LAT1 in grass carp (Ctenopharyngodon idellus) is currently unknown. In the present study, full-length cDNA (2,688 bp), which encodes y⁺LAT1 and contains a 5'-untranslated region (319 bp), an open reading frame (1,506 bp) and a 3'-untranslated region (863 bp), has been cloned from grass carp. Amino acid sequence of grass carp y⁺LAT1 contains 11 transmembrane domains and shows 95 %, 80 % and 75 % sequence similarity to zebra fish, amphibian and mammalian y⁺LAT1, respectively. The tissue distribution and expression regulation by fasting of y⁺LAT1 mRNA were analyzed using real-time PCR. Our results showed that y⁺LAT1 mRNA was highly expressed in midgut, foregut and spleen while weakly expressed in hindgut, kidney, gill, brain, heart, liver and muscle. Nutritional status significantly influenced y⁺LAT1 mRNA expression in fish tissues, such as down-regulation of y⁺LAT1 mRNA expression after fasting (14 days).
Collapse
Affiliation(s)
- Jixuan Yang
- Fisheries College, Huazhong Agricultural University, Wuhan, 430070, China
| | | | | | | | | | | |
Collapse
|
32
|
Chen RF, Wang L, Cheng JT, Yang KD. Induction of IFNα or IL-12 depends on differentiation of THP-1 cells in dengue infections without and with antibody enhancement. BMC Infect Dis 2012; 12:340. [PMID: 23216989 PMCID: PMC3575308 DOI: 10.1186/1471-2334-12-340] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 12/04/2012] [Indexed: 12/22/2022] Open
Abstract
Background Appropriate induction of the early Th1 cytokine IL-12 is a critical defense directed against viral infection. We have previously shown that different viruses elicited either IL-12 or IFNα dependent Th1 reactions. Using dengue-2 virus, we sought to explore how dengue-2 induced IL-12 or IFNα expression by monocytic and its derived dendritic cells. Methods We employed human monocytic cell line, THP-1, to investigate whether differentiation of monocytic cells is involved in the switch between IFNα and IL-12 induction. Flow cytometry, RT-PCR and ELISA were respectively used to determine cell differentiation, IL-12 and IFNα mRNA expression and protein production. Results THP-1, expressing CD123, which is a plasmacytoid dendritic cell marker, but not CD14, CD11b or CD11c revealed IFNα mRNA expression while stimulated by dengue-2. In contrast, PMA-induced THP-1 differentiation toward monocytic cells expressed CD11b+, and CD14+, but not CD123, and revealed exclusively IL-12 expression while stimulated by dengue-2. Further studies showed that CD123+ expressing THP-1 cells elicited higher IFNα expression in dose and time dependent induction after infection, and PMA-induced monocytic differentiation of THP-1 cells revealed IL-12 expression. Antibody-dependent enhancement of DEN-2 infection significantly suppressed the DEN-2 induced IL-12 p40 expression in monocytic differentiated THP-1 cells. Conclusions Clarification and modulation of the early Th1 reaction in different monocytic cells may change or prevent complication from dengue infection.
Collapse
Affiliation(s)
- Rong-Fu Chen
- Department of Medical Research and Development, Show Chwan Health Care System, Changhua, Taiwan
| | | | | | | |
Collapse
|
33
|
Abstract
Nitric oxide (NO) is an endogenous vasodilator molecule synthetized from L-arginine by a family of nitric oxide synthases. In differentiated human endothelial cells, it is well known that L-arginine uptake via cationic amino acid transporters (y(+)/CAT) or system y(+)L is required for the NO synthesis via endothelial nitric oxide synthase, but there are no reports in human endothelial progenitor cell (hEPC). Therefore, we isolated hEPCs from peripheral blood of healthy donors and cultured them for either 3 (hEPC-3d) or 14 days (hEPC-14d) to characterize the L-arginine transport and NO synthesis in those cells. L-arginine transport and NO synthesis were analyzed in the presence or absence of N-ethylmaleimide or L-nitroarginine methyl ester, as inhibitors of y(+)/CAT system and nitric oxide synthases, respectively. The results showed that L-arginine uptake is higher in hEPC-14d than in hEPC-3d. Kinetic parameters for L-arginine transport showed the existence of at least 2 transporter systems in hEPC: a high affinity transporter system (K(m)= 4.8 ± 1.1 μM for hEPC-3d and 6.1 ± 2.4 μM for hEPC-14d) and a medium affinity transporter system (K(m) = 85.1 ± 4.0 μM for hEPC-3d and 95.1 ± 8 μM for hEPC-14d). Accordingly, hEPC expressed mRNA and protein for CAT-1 (ie, system y(+)) and mRNA for 2 subunits of y(+)L system, yLAT1, and 4F2hc. Higher L-citruline production and NO bioavailability (4-fold), and endothelial nitric oxide synthase expression (both mRNA and protein) were observed in hEPC-14d compared with hEPC-3d. Finally, the high L-citruline formation observed in hEPC-14d was blocked by N-ethylmaleimide. In conclusion, this study allowed to identity a functional L-arginine/NO pathway in two hEPC differentiation stages, which improves the understanding of the physiology of these precursor cells.
Collapse
|
34
|
|
35
|
Tringham M, Kurko J, Tanner L, Tuikkala J, Nevalainen OS, Niinikoski H, Näntö-Salonen K, Hietala M, Simell O, Mykkänen J. Exploring the transcriptomic variation caused by the Finnish founder mutation of lysinuric protein intolerance (LPI). Mol Genet Metab 2012; 105:408-15. [PMID: 22221392 DOI: 10.1016/j.ymgme.2011.12.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 12/09/2011] [Indexed: 12/19/2022]
Abstract
Lysinuric protein intolerance (LPI) is an autosomal recessive disorder caused by mutations in cationic amino acid transporter gene SLC7A7. Although all Finnish patients share the same homozygous mutation, their clinical manifestations vary greatly. The symptoms range from failure to thrive, protein aversion, anemia and hyperammonaemia, to immunological abnormalities, nephropathy and pulmonary alveolar proteinosis. To unravel the molecular mechanisms behind those symptoms not explained directly by the primary mutation, gene expression profiles of LPI patients were studied using genome-wide microarray technology. As a result, we discovered 926 differentially-expressed genes, including cationic and neutral amino acid transporters. The functional annotation analysis revealed a significant accumulation of such biological processes as inflammatory response, immune system processes and apoptosis. We conclude that changes in the expression of genes other than SLC7A7 may be linked to the various symptoms of LPI, indicating a complex interplay between amino acid transporters and various cellular processes.
Collapse
Affiliation(s)
- Maaria Tringham
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|