151
|
Ekuban A, Shichino S, Zong C, Ekuban FA, Kinoshita K, Ichihara S, Matsushima K, Ichihara G. Transcriptome analysis of human cholangiocytes exposed to carcinogenic 1,2-dichloropropane in the presence of macrophages in vitro. Sci Rep 2022; 12:11222. [PMID: 35780190 PMCID: PMC9250500 DOI: 10.1038/s41598-022-15295-3] [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: 03/18/2022] [Accepted: 06/22/2022] [Indexed: 11/09/2022] Open
Abstract
1,2-Dichloropropane (1,2-DCP), a synthetic organic solvent, has been implicated in causality of cholangiocarcinoma (bile duct cancer). 1,2-DCP-induced occupational cholangiocarcinoma show a different carcinogenic process compared to common cholangiocarcinoma, but its mechanism remains elusive. We reported previously that exposure of MMNK-1 cholangiocytes co-cultured with THP-1 macrophages, but not monocultured MMNK-1 cholangiocytes, to 1,2-DCP induced activation-induced cytidine deaminase (AID) expression, DNA damage and ROS production. The aim of this study was to identify relevant biological processes or target genes expressed in response to 1,2-DCP, using an in vitro system where cholangiocytes are co-cultured with macrophages. The co-cultured cells were exposed to 1,2-DCP at 0, 0.1 or 0.4 mM for 24 h, and then the cell lysates were assessed by transcriptome analysis. 1,2-DCP upregulated the expression of base excision repair genes in MMNK-1 cholangiocytes in the co-cultures, whereas it upregulated the expression of cell cycle-related genes in THP-1 macrophages. Activation of the base excision repair pathway might result from the previously observed DNA damage in MMNK-1 cholangiocytes co-cultured with THP-1 macrophages, although involvement of other mechanisms such as DNA replication, cell death or other types of DNA repair was not disproved. Cross talk interactions between cholangiocytes and macrophages leading to DNA damage in the cholangiocytes should be explored.
Collapse
Affiliation(s)
- Abigail Ekuban
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Building No. 15, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda, 278-0022, Japan
| | - Cai Zong
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Building No. 15, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Frederick Adams Ekuban
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Building No. 15, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kazuo Kinoshita
- Evolutionary Medicine, Shizuoka Graduate University of Public Health, Shizuoka, 420-0881, Japan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke, 329-0498, Japan
| | - Kouji Matsushima
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda, 278-0022, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Building No. 15, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan.
| |
Collapse
|
152
|
Transcriptome Analysis of Intracellular Amastigotes of Clinical Leishmania infantum Lines from Therapeutic Failure Patients after Infection of Human Macrophages. Microorganisms 2022; 10:microorganisms10071304. [PMID: 35889023 PMCID: PMC9324091 DOI: 10.3390/microorganisms10071304] [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: 05/17/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/23/2022] Open
Abstract
Leishmaniasis is considered to be one of the most neglected tropical diseases affecting humans and animals around the world. Due to the absence of an effective vaccine, current treatment is based on chemotherapy. However, the continuous appearance of drug resistance and therapeutic failure (TF) lead to an early obsolescence of treatments. Identification of the factors that contribute to TF and drug resistance in leishmaniasis will constitute a useful tool for establishing future strategies to control this disease. In this manuscript, we evaluated the transcriptomic changes in the intracellular amastigotes of the Leishmania infantum parasites isolated from patients with leishmaniasis and TF at 96 h post-infection of THP-1 cells. The adaptation of the parasites to their new environment leads to expression alterations in the genes involved mainly in the transport through cell membranes, energy and redox metabolism, and detoxification. Specifically, the gene that codes for the prostaglandin f2α synthase seems to be relevant in the pathogenicity and TF since it appears substantially upregulated in all the L. infantum lines. Overall, our results show that at the late infection timepoint, the transcriptome of the parasites undergoes significant changes that probably improve the survival of the Leishmania lines in the host cells, contributing to the TF phenotype as well as drug therapy evasion.
Collapse
|
153
|
Kim SM, Studnitzer B, Esser-Kahn A. Heat Shock Protein 90's Mechanistic Role in Contact Hypersensitivity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2622-2631. [PMID: 35675957 PMCID: PMC9308677 DOI: 10.4049/jimmunol.2101023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/15/2022] [Indexed: 11/19/2022]
Abstract
Despite the known dangers of contact allergens and their long-lasting use as models in immunology, their molecular mode of action largely remains unknown. In this study, we report that a contact allergen, 1-chloro-2,4-dinitrobenzene (DNCB), elicits contact hypersensitivity through binding the protein we identify. Starting from an unbiased sampling of proteomics, we found nine candidate proteins with unique DNCB-modified peptide fragments. More than half of these fragments belonged to heat shock protein 90 (HSP90), a common stress-response protein and a damage-associated molecular pattern, and showed the highest probability of incidence. Inhibition and short hairpin RNA knockdown of HSP90 in human monocyte cell line THP-1 suppressed the potency of DNCB by >80%. Next, we successfully reduced DNCB-induced contact hypersensitivity in HSP90-knockout mice, which confirmed our findings. Finally, we hypothesized that DNCB-modified HSP90 activates the immune cells through HSP90's receptor, CD91. Pretreatment of CD91 in THP-1 cell lines and BALB/c mice attenuated the potency of DNCB, consistent with the result of HSP90-knockout mice. Altogether, our data show that DNCB-HSP90 binding plays a role in mediating DNCB-induced contact hypersensitivity, and the activation of CD91 by DNCB-modified HSP90 proteins could mediate this process.
Collapse
Affiliation(s)
- Seong-Min Kim
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL
| | - Bradley Studnitzer
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL
| | - Aaron Esser-Kahn
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL
| |
Collapse
|
154
|
Comparing α-Quartz-Induced Cytotoxicity and Interleukin-8 Release in Pulmonary Mono- and Co-Cultures Exposed under Submerged and Air-Liquid Interface Conditions. Int J Mol Sci 2022; 23:ijms23126412. [PMID: 35742856 PMCID: PMC9224477 DOI: 10.3390/ijms23126412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 02/06/2023] Open
Abstract
The occupational exposure to particles such as crystalline quartz and its impact on the respiratory tract have been studied extensively in recent years. For hazard assessment, the development of physiologically more relevant in-vitro models, i.e., air-liquid interface (ALI) cell cultures, has greatly progressed. Within this study, pulmonary culture models employing A549 and differentiated THP-1 cells as mono-and co-cultures were investigated. The different cultures were exposed to α-quartz particles (Min-U-Sil5) with doses ranging from 15 to 66 µg/cm2 under submerged and ALI conditions and cytotoxicity as well as cytokine release were analyzed. No cytotoxicity was observed after ALI exposure. Contrarily, Min-U-Sil5 was cytotoxic at the highest dose in both submerged mono- and co-cultures. A concentration-dependent release of interleukin-8 was shown for both exposure types, which was overall stronger in co-cultures. Our findings showed considerable differences in the toxicological responses between ALI and submerged exposure and between mono- and co-cultures. A substantial influence of the presence or absence of serum in cell culture media was noted as well. Within this study, the submerged culture was revealed to be more sensitive. This shows the importance of considering different culture and exposure models and highlights the relevance of communication between different cell types for toxicological investigations.
Collapse
|
155
|
Kimiz‐Gebologlu I, Demirden SF, Oncel SS. A study of the THP‐1 cell line as the potential biologics production platform with the emphasis on serum‐free media substitution for economic expediency. Biotechnol J 2022; 17:e2200154. [DOI: 10.1002/biot.202200154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 11/09/2022]
Affiliation(s)
| | - S. Furkan Demirden
- Department of Bioengineering Faculty of Engineering Ege University Izmir Turkiye
| | - Suphi S. Oncel
- Department of Bioengineering Faculty of Engineering Ege University Izmir Turkiye
| |
Collapse
|
156
|
HIV Latency in Myeloid Cells: Challenges for a Cure. Pathogens 2022; 11:pathogens11060611. [PMID: 35745465 PMCID: PMC9230125 DOI: 10.3390/pathogens11060611] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/10/2022] [Accepted: 05/21/2022] [Indexed: 01/27/2023] Open
Abstract
The use of antiretroviral therapy (ART) for Human Immunodeficiency Virus (HIV) treatment has been highly successful in controlling plasma viremia to undetectable levels. However, a complete cure for HIV is hindered by the presence of replication-competent HIV, integrated in the host genome, that can persist long term in a resting state called viral latency. Resting memory CD4+ T cells are considered the biggest reservoir of persistent HIV infection and are often studied exclusively as the main target for an HIV cure. However, other cell types, such as circulating monocytes and tissue-resident macrophages, can harbor integrated, replication-competent HIV. To develop a cure for HIV, focus is needed not only on the T cell compartment, but also on these myeloid reservoirs of persistent HIV infection. In this review, we summarize their importance when designing HIV cure strategies and challenges associated to their identification and specific targeting by the “shock and kill” approach.
Collapse
|
157
|
Akula S, Lara S, Olsson AK, Hellman L. Quantitative Analysis of the Transcriptome of Two Commonly Used Human Monocytic Cell Lines—THP-1 and Mono Mac 6—Reveals Their Arrest during Early Monocyte/Neutrophil Differentiation. Int J Mol Sci 2022; 23:ijms23105818. [PMID: 35628628 PMCID: PMC9145822 DOI: 10.3390/ijms23105818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 12/10/2022] Open
Abstract
Cell lines of monocyte/macrophage origin are often used as model systems to study monocyte/macrophage biology. A relevant question is how similar these cell lines are to their in vivo counterparts? To address this issue, we performed a detailed analysis of the transcriptome of two commonly used human monocyte/macrophage cell lines, Mono Mac 6 and THP-1. Both of these cell lines originate from leukemic cells with myelo-monocytic characteristics. We found that both Mono Mac 6 and THP-1 represent cells of very immature origin. Their transcriptomes show more similarities to immature neutrophils than cells of the monocyte/macrophage lineage. They express significant levels of N-elastase, proteinase 3, cathepsin G, and azurocidin but very low levels of CD14, ficolin, and complement factor P. All major MHC class II genes are also expressed at low levels. They show high levels of lysozyme and low levels of one of the immunoglobulin Fc receptors, FCGRIIA, which is characteristic of both neutrophils and monocytes. THP-1, but not Mono Mac 6, also expresses the high-affinity receptor for IgG, FCGRIA. Both cell lines lack the expression of the connective tissue components fibronectin, proteoglycan 4, and syndecan 3, which are characteristics of tissue macrophages but are absent in blood monocytes, indicating that they originate from bone marrow precursors and not yolk sac-derived hematopoietic cells. Both of these cell lines seem, therefore, to represent cells arrested during early myelo-monocytic development, at a branch point between neutrophil and monocyte differentiation. Their very immature phenotype indicates that great care should be taken when using these cell lines as models for normal monocyte/macrophage biology.
Collapse
Affiliation(s)
- Srinivas Akula
- Department of Cell and Molecular Biology, Uppsala University, The Biomedical Center, P.O. Box 596, SE-75124 Uppsala, Sweden
- Department of Anatomy, Physiology, and Biochemistry, Swedish University of Agricultural Sciences, Box 7011, SE-75007 Uppsala, Sweden
| | - Sandra Lara
- Department of Cell and Molecular Biology, Uppsala University, The Biomedical Center, P.O. Box 596, SE-75124 Uppsala, Sweden
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, BMC, Box 582, SE-75123 Uppsala, Sweden
| | - Lars Hellman
- Department of Cell and Molecular Biology, Uppsala University, The Biomedical Center, P.O. Box 596, SE-75124 Uppsala, Sweden
| |
Collapse
|
158
|
Su S, Lei A, Wang X, Lu H, Wang S, Yang Y, Li N, Zhang Y, Zhang J. Induced CAR-Macrophages as a Novel Therapeutic Cell Type for Cancer Immune Cell Therapies. Cells 2022; 11:1652. [PMID: 35626689 PMCID: PMC9139529 DOI: 10.3390/cells11101652] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/09/2022] [Accepted: 04/22/2022] [Indexed: 01/27/2023] Open
Abstract
The Chimeric antigen receptor (CAR)-T cell therapy has made inroads in treating hematological malignancies. Nonetheless, there are still multiple hurdles in CAR-T cell therapy for solid tumors. Primary CAR-expressing macrophage cells (CAR-Ms) and induced pluripotent stem cells (iPSCs)-derived CAR-expressing macrophage cells (CAR-iMacs) have emerged as attractive alternatives in our quest for an efficient and inexpensive approach for tumor immune cell therapy. In this review, we list the current state of development of human CAR-macrophages and provide an overview of the crucial functions of human CAR-macrophages in the field of tumor immune cell therapy.
Collapse
Affiliation(s)
- Siyu Su
- Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China;
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China; (A.L.); (X.W.); (H.L.)
| | - Anhua Lei
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China; (A.L.); (X.W.); (H.L.)
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou 310058, China
| | - Xudong Wang
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China; (A.L.); (X.W.); (H.L.)
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou 310058, China
| | - Hengxing Lu
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China; (A.L.); (X.W.); (H.L.)
| | - Shuhang Wang
- National Cancer Center/National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Beijing 100021, China; (S.W.); (N.L.)
| | - Yuqi Yang
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People’s Hospital, No. 83 Zhongshan Road, Guiyang 550000, China;
| | - Ning Li
- National Cancer Center/National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Beijing 100021, China; (S.W.); (N.L.)
| | - Yi Zhang
- Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China;
| | - Jin Zhang
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China; (A.L.); (X.W.); (H.L.)
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
159
|
Doi H, Matsui T, Dijkstra JM, Ogasawara A, Higashimoto Y, Imamura S, Ohye T, Takematsu H, Katsuda I, Akiyama H. Andrographolide, isolated from Andrographis paniculata, induces apoptosis in monocytic leukemia and multiple myeloma cells via augmentation of reactive oxygen species production. F1000Res 2022; 10:542. [PMID: 35528957 PMCID: PMC9069414 DOI: 10.12688/f1000research.53595.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/20/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Andrographolide (Andro) is a diterpenoid component of the plant
Andrographis paniculata that is known for its anti-tumor activity against a variety of cancer cells. Methods: We studied the effects of Andro on the viability of the human leukemia monocytic cell line THP-1 and the human multiple myeloma cell line H929. Andro was compared with cytosine arabinoside (Ara-C) and vincristine (VCR), which are well-established therapeutics against hematopoietic tumors. The importance of reactive oxygen species (ROS) production for the toxicity of each agent was investigated by using an inhibitor of ROS production, N-acetyl-L-cysteine (NAC). Results: Andro reduced the viability of THP-1 and H929 in a concentration-dependent manner. H929 viability was highly susceptible to Andro, although only slightly susceptible to Ara-C. The agents Andro, Ara-C, and VCR each induced apoptosis, as shown by cellular shrinkage, DNA fragmentation, and increases in annexin V-binding, caspase-3/7 activity, ROS production, and mitochondrial membrane depolarization. Whereas Ara-C and VCR increased the percentages of cells in the G0/G1 and G2/M phases, respectively, Andro showed little or no detectable effect on cell cycle progression. The apoptotic activities of Andro were largely suppressed by NAC, an inhibitor of ROS production, whereas NAC hardly affected the apoptotic activities of Ara-C and VCR. Conclusions: Andro induces ROS-dependent apoptosis in monocytic leukemia THP-1 and multiple myeloma H929 cells, underlining its potential as a therapeutic agent for treating hematopoietic tumors. The high toxicity for H929 cells, by a mechanism that is different from that of Ara-C and VCR, is encouraging for further studies on the use of Andro against multiple myeloma.
Collapse
Affiliation(s)
- Hiroki Doi
- Field of Clinical Laboratory Sciences, Fujita Health University Graduate School of Health Sciences, Toyoake, 470-1192, Japan
| | - Taei Matsui
- Field of Clinical Laboratory Sciences, Fujita Health University Graduate School of Health Sciences, Toyoake, 470-1192, Japan
- Faculty of Medical Technology, Fujita Health University School of Medical Sciences, Toyoake, 470-1192, Japan
| | - Johannes M. Dijkstra
- Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, 470-1192, Japan
| | - Atsushi Ogasawara
- Field of Clinical Laboratory Sciences, Fujita Health University Graduate School of Health Sciences, Toyoake, 470-1192, Japan
| | - Yuki Higashimoto
- Faculty of Medical Technology, Fujita Health University School of Medical Sciences, Toyoake, 470-1192, Japan
| | - Seiji Imamura
- Faculty of Medical Technology, Fujita Health University School of Medical Sciences, Toyoake, 470-1192, Japan
| | - Tamae Ohye
- Faculty of Medical Technology, Fujita Health University School of Medical Sciences, Toyoake, 470-1192, Japan
| | - Hiromu Takematsu
- Field of Clinical Laboratory Sciences, Fujita Health University Graduate School of Health Sciences, Toyoake, 470-1192, Japan
- Faculty of Medical Technology, Fujita Health University School of Medical Sciences, Toyoake, 470-1192, Japan
| | - Itsuro Katsuda
- Department of Hematology, Fujita Health University School of Medicine, Toyoake, 470-1192, Japan
| | - Hidehiko Akiyama
- Field of Clinical Laboratory Sciences, Fujita Health University Graduate School of Health Sciences, Toyoake, 470-1192, Japan
- Faculty of Medical Technology, Fujita Health University School of Medical Sciences, Toyoake, 470-1192, Japan
| |
Collapse
|
160
|
Serrero MC, Girault V, Weigang S, Greco TM, Ramos-Nascimento A, Anderson F, Piras A, Hickford Martinez A, Hertzog J, Binz A, Pohlmann A, Prank U, Rehwinkel J, Bauerfeind R, Cristea IM, Pichlmair A, Kochs G, Sodeik B. The interferon-inducible GTPase MxB promotes capsid disassembly and genome release of herpesviruses. eLife 2022; 11:e76804. [PMID: 35475759 PMCID: PMC9150894 DOI: 10.7554/elife.76804] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/22/2022] [Indexed: 11/18/2022] Open
Abstract
Host proteins sense viral products and induce defence mechanisms, particularly in immune cells. Using cell-free assays and quantitative mass spectrometry, we determined the interactome of capsid-host protein complexes of herpes simplex virus and identified the large dynamin-like GTPase myxovirus resistance protein B (MxB) as an interferon-inducible protein interacting with capsids. Electron microscopy analyses showed that cytosols containing MxB had the remarkable capability to disassemble the icosahedral capsids of herpes simplex viruses and varicella zoster virus into flat sheets of connected triangular faces. In contrast, capsids remained intact in cytosols with MxB mutants unable to hydrolyse GTP or to dimerize. Our data suggest that MxB senses herpesviral capsids, mediates their disassembly, and thereby restricts the efficiency of nuclear targeting of incoming capsids and/or the assembly of progeny capsids. The resulting premature release of viral genomes from capsids may enhance the activation of DNA sensors, and thereby amplify the innate immune responses.
Collapse
Affiliation(s)
- Manutea C Serrero
- Institute of Virology, Hannover Medical SchoolHannoverGermany
- RESIST - Cluster of Excellence, Hannover Medical SchoolHannoverGermany
| | | | - Sebastian Weigang
- Institute of Virology, Freiburg University Medical Center, University of FreiburgFreiburgGermany
| | - Todd M Greco
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | | | - Fenja Anderson
- Institute of Virology, Hannover Medical SchoolHannoverGermany
| | - Antonio Piras
- Institute of Virology, Technical University MunichMunichGermany
| | | | - Jonny Hertzog
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Anne Binz
- Institute of Virology, Hannover Medical SchoolHannoverGermany
- RESIST - Cluster of Excellence, Hannover Medical SchoolHannoverGermany
- German Center for Infection Research (DZIF), Hannover-Braunschweig Partner SiteHannoverGermany
| | - Anja Pohlmann
- Institute of Virology, Hannover Medical SchoolHannoverGermany
- RESIST - Cluster of Excellence, Hannover Medical SchoolHannoverGermany
- German Center for Infection Research (DZIF), Hannover-Braunschweig Partner SiteHannoverGermany
| | - Ute Prank
- Institute of Virology, Hannover Medical SchoolHannoverGermany
| | - Jan Rehwinkel
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Rudolf Bauerfeind
- Research Core Unit Laser Microscopy, Hannover Medical SchoolHannoverGermany
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Andreas Pichlmair
- Institute of Virology, Technical University MunichMunichGermany
- German Center for Infection Research (DZIF), Munich Partner siteMunichGermany
| | - Georg Kochs
- Institute of Virology, Freiburg University Medical Center, University of FreiburgFreiburgGermany
| | - Beate Sodeik
- Institute of Virology, Hannover Medical SchoolHannoverGermany
- RESIST - Cluster of Excellence, Hannover Medical SchoolHannoverGermany
- German Center for Infection Research (DZIF), Hannover-Braunschweig Partner SiteHannoverGermany
| |
Collapse
|
161
|
Joshi A, Soni A, Acharya S. In vitro models and ex vivo systems used in inflammatory bowel disease. IN VITRO MODELS 2022; 1:213-227. [PMID: 37519330 PMCID: PMC9036838 DOI: 10.1007/s44164-022-00017-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 12/15/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic, relapsing gastrointestinal condition. Ulcerative colitis and Crohn's disease are types of inflammatory bowel disease. Over many decades, the disease has been a topic of study, with experts still trying to figure out its cause and pathology. Researchers have established many in vivo animal models, in vitro cell lines, and ex vivo systems to understand its cause ultimately and adequately identify a therapy. However, in vivo animal models cannot be regarded as good models for studying IBD since they cannot completely simulate the disease. Furthermore, because species differences are a crucial subject of concern, in vitro cell lines and ex vivo systems can be employed to recreate the condition properly. In vitro models serve as the starting point for biological and medical research. Ex vivo and in vitro models for replicating gut physiology have been developed. This review aims to present a clear understanding of several in vitro and ex vivo models of IBD and provide insights into their benefits and limits and their value in understanding intestinal physiology.
Collapse
Affiliation(s)
- Abhishek Joshi
- Department of Pharmacology, SSR College of Pharmacy, Union Territory of Dadra 396230 Sayli, Silvassa, India
| | - Arun Soni
- Department of Pharmacology, SSR College of Pharmacy, Union Territory of Dadra 396230 Sayli, Silvassa, India
| | - Sanjeev Acharya
- Department of Pharmacognosy, SSR College of Pharmacy, Union Territory of Dadra 396230 Sayli, Silvassa, India
| |
Collapse
|
162
|
Kasai F, Hirayama N, Fukushima M, Kohara A, Nakamura Y. THP
‐1 reference data: proposal of an
in vitro
branched evolution model for cancer cell lines. Int J Cancer 2022; 151:463-472. [DOI: 10.1002/ijc.34019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 03/09/2022] [Accepted: 03/22/2022] [Indexed: 11/12/2022]
Affiliation(s)
- Fumio Kasai
- RIKEN Cell Bank, Cell Engineering Division, BioResource Research Center Tsukuba Japan
- JCRB Cell Bank, National Institutes of Biomedical Innovation, Health and Nutrition Osaka Japan
| | - Noriko Hirayama
- JCRB Cell Bank, National Institutes of Biomedical Innovation, Health and Nutrition Osaka Japan
| | - Makoto Fukushima
- RIKEN Cell Bank, Cell Engineering Division, BioResource Research Center Tsukuba Japan
| | - Arihiro Kohara
- JCRB Cell Bank, National Institutes of Biomedical Innovation, Health and Nutrition Osaka Japan
| | - Yukio Nakamura
- RIKEN Cell Bank, Cell Engineering Division, BioResource Research Center Tsukuba Japan
| |
Collapse
|
163
|
Valestrand L, Zheng F, Hansen SH, Øgaard J, Hov JR, Björkström NK, Karlsen TH, Jiang X, Melum E. Bile from Patients with Primary Sclerosing Cholangitis Contains Mucosal-Associated Invariant T-Cell Antigens. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:629-641. [PMID: 35063408 DOI: 10.1016/j.ajpath.2021.12.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/23/2021] [Accepted: 12/16/2021] [Indexed: 12/14/2022]
Abstract
Primary sclerosing cholangitis (PSC) is associated with altered microbiota of the gut and bile. Mucosal-associated invariant T (MAIT) cells, enriched in human liver, uniquely recognize microbial-derived metabolites. This study aimed to determine whether bile from patients with PSC contains antigens activating MAIT cells. Bile was collected at the time of liver transplantation from patients with PSC (n = 28). The bile samples were either directly incubated with peripheral blood mononuclear cells from healthy donors or with antigen-presenting cells followed by co-culture with peripheral blood mononuclear cells. MAIT cell activation was assessed by flow cytometry. An anti-MR1 antibody was used to determine whether the activation was major histocompatibility complex class I-related protein (MR1) restricted. Biliary microbiota profiles were generated using 16S rRNA amplicon sequencing, and the abundance of the bacterial gene ribD was predicted. Eight of 28 bile samples could activate MAIT cells. This activation was partly MR1-dependent in five of eight bile samples. Microbial DNA was detected in 15 of 28 bile samples, including the five bile samples leading to MR1-dependent activation. A higher abundance of the ribD gene expression in the group of bile samples that could activate MAIT cells was predicted on the basis of the 16S sequencing. In co-culture experiments, cholangiocytes could take up and present biliary antigens to MAIT cells. These findings suggest a pathophysiological pathway in PSC connecting the immune system and the microbiome.
Collapse
Affiliation(s)
- Laura Valestrand
- Norwegian PSC Research Center, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section of Gastroenterology, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway
| | - Fei Zheng
- Norwegian PSC Research Center, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Simen H Hansen
- Norwegian PSC Research Center, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Jonas Øgaard
- Norwegian PSC Research Center, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Johannes R Hov
- Norwegian PSC Research Center, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section of Gastroenterology, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Tom H Karlsen
- Norwegian PSC Research Center, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section of Gastroenterology, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway
| | - Xiaojun Jiang
- Norwegian PSC Research Center, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen Melum
- Norwegian PSC Research Center, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section of Gastroenterology, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway; Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
164
|
Duweb A, Gaiser AK, Stiltz I, El Gaafary M, Simmet T, Syrovets T. The SC cell line as an in vitro model of human monocytes. J Leukoc Biol 2022; 112:659-668. [PMID: 35355303 DOI: 10.1002/jlb.1a1221-680r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/01/2022] [Indexed: 11/08/2022] Open
Abstract
In vitro analysis of human macrophages is generally hampered by the necessity to differentiate them from peripheral blood monocytes. We have analyzed to which extent noncancerous SC monocytes could be used as an in vitro macrophage model. Macrophages differentiated from peripheral monocytes using standard CSF1 and CSF2 protocols for M2 and M1 precursors, respectively, were compared with THP-1-derived macrophages treated with PMA and with SC-derived macrophages differentiated either by CSF1, CSF2, or PMA according to different protocols. The optimal condition for generation of SC macrophages was treatment with PMA for 3 days, followed by 5-days culture without PMA and 24-h polarization with LPS/IFN-γ or IL-4/IL-13. Similar to THP-1, SC cells do not express the monocyte marker CD14 and differentiation to macrophages results neither in CD68 nor in CD14 expression, both of which were expressed by monocyte-derived macrophages. Similar to THP-1-macrophages, a proportion of SC macrophages can be polarized to the M1-like subtype that is characterized by higher expression of CD38, CD86, CD80, TNF-α, and IL-1ra, whereas treatment with IL4/IL13 did not lead to expression of the M2-associated receptors CD163, CD206, and only slightly increased the CD200R expression. Still, SC-M1 express much lower levels of the M1-associated markers compared with monocyte-derived M1 and no IL-1β. The data demonstrate that SC-derived macrophages differ from monocyte-derived macrophages in respect of their morphology, expression of important macrophage markers, phagocytosis. Yet, polarized SC-M1-like cells may with restrictions serve as a model for M1 macrophages, though this model does not provide significant advantages over already well-described THP-1-M1-like cells.
Collapse
Affiliation(s)
- Amira Duweb
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany.,Department of Pharmacology, Faculty of Medicine, University of Tripoli, Tripoli, Libya
| | - Ann-Kathrin Gaiser
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Isabel Stiltz
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Menna El Gaafary
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany.,Department of Pharmacognosy, College of Pharmacy, Cairo University, Cairo, Egypt
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Tatiana Syrovets
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| |
Collapse
|
165
|
Robida PA, Rische CH, Morgenstern NBB, Janarthanam R, Cao Y, Krier-Burris RA, Korver W, Xu A, Luu T, Schanin J, Leung J, Rothenberg ME, Wechsler JB, Youngblood BA, Bochner BS, O’Sullivan JA. Functional and Phenotypic Characterization of Siglec-6 on Human Mast Cells. Cells 2022; 11:1138. [PMID: 35406705 PMCID: PMC8997871 DOI: 10.3390/cells11071138] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
Mast cells are tissue-resident cells that contribute to allergic diseases, among others, due to excessive or inappropriate cellular activation and degranulation. Therapeutic approaches to modulate mast cell activation are urgently needed. Siglec-6 is an immunoreceptor tyrosine-based inhibitory motif (ITIM)-bearing receptor selectively expressed by mast cells, making it a promising target for therapeutic intervention. However, the effects of its engagement on mast cells are poorly defined. Siglec-6 expression and endocytosis on primary human mast cells and mast cell lines were assessed by flow cytometry. SIGLEC6 mRNA expression was examined by single-cell RNAseq in esophageal tissue biopsy samples. The ability of Siglec-6 engagement or co-engagement to prevent primary mast cell activation was determined based on assessments of mediator and cytokine secretion and degranulation markers. Siglec-6 was highly expressed by all mast cells examined, and the SIGLEC6 transcript was restricted to mast cells in esophageal biopsy samples. Siglec-6 endocytosis occurred with delayed kinetics relative to the related receptor Siglec-8. Co-crosslinking of Siglec-6 with FcεRIα enhanced the inhibition of mast cell activation and diminished downstream ERK1/2 and p38 phosphorylation. The selective, stable expression and potent inhibitory capacity of Siglec-6 on human mast cells are favorable for its use as a therapeutic target in mast cell-driven diseases.
Collapse
Affiliation(s)
- Piper A. Robida
- Division of Allergy and Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (P.A.R.); (Y.C.); (R.A.K.-B.); (J.B.W.); (B.S.B.)
| | - Clayton H. Rische
- McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA;
| | - Netali Ben-Baruch Morgenstern
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; (N.B.-B.M.); (M.E.R.)
| | - Rethavathi Janarthanam
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA;
| | - Yun Cao
- Division of Allergy and Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (P.A.R.); (Y.C.); (R.A.K.-B.); (J.B.W.); (B.S.B.)
| | - Rebecca A. Krier-Burris
- Division of Allergy and Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (P.A.R.); (Y.C.); (R.A.K.-B.); (J.B.W.); (B.S.B.)
| | - Wouter Korver
- Allakos, Inc., Redwood City, CA 94065, USA; (W.K.); (A.X.); (T.L.); (J.S.); (J.L.); (B.A.Y.)
| | - Alan Xu
- Allakos, Inc., Redwood City, CA 94065, USA; (W.K.); (A.X.); (T.L.); (J.S.); (J.L.); (B.A.Y.)
| | - Thuy Luu
- Allakos, Inc., Redwood City, CA 94065, USA; (W.K.); (A.X.); (T.L.); (J.S.); (J.L.); (B.A.Y.)
| | - Julia Schanin
- Allakos, Inc., Redwood City, CA 94065, USA; (W.K.); (A.X.); (T.L.); (J.S.); (J.L.); (B.A.Y.)
| | - John Leung
- Allakos, Inc., Redwood City, CA 94065, USA; (W.K.); (A.X.); (T.L.); (J.S.); (J.L.); (B.A.Y.)
| | - Marc E. Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; (N.B.-B.M.); (M.E.R.)
| | - Joshua B. Wechsler
- Division of Allergy and Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (P.A.R.); (Y.C.); (R.A.K.-B.); (J.B.W.); (B.S.B.)
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA;
| | - Bradford A. Youngblood
- Allakos, Inc., Redwood City, CA 94065, USA; (W.K.); (A.X.); (T.L.); (J.S.); (J.L.); (B.A.Y.)
| | - Bruce S. Bochner
- Division of Allergy and Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (P.A.R.); (Y.C.); (R.A.K.-B.); (J.B.W.); (B.S.B.)
| | - Jeremy A. O’Sullivan
- Division of Allergy and Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (P.A.R.); (Y.C.); (R.A.K.-B.); (J.B.W.); (B.S.B.)
| |
Collapse
|
166
|
Vitamin D Counteracts Lipid Accumulation, Augments Free Fatty Acid-Induced ABCA1 and CPT-1A Expression While Reducing CD36 and C/EBPβ Protein Levels in Monocyte-Derived Macrophages. Biomedicines 2022; 10:biomedicines10040775. [PMID: 35453525 PMCID: PMC9028184 DOI: 10.3390/biomedicines10040775] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/16/2022] [Accepted: 03/25/2022] [Indexed: 12/15/2022] Open
Abstract
The biologically active form of vitamin D, calcitriol (VD3), has received great attention for its extraskeletal effects, such as a protective role on the cardiovascular system. The aim of the present work is to test the capacity of VD3 to affect lipid metabolism and fatty acid accumulation in an in vitro model of monocyte (THP-1)-derived macrophages. Cells were treated for 24 h with oleic/palmitic acid (500 μM, 2:1 ratio) and different VD3 concentrations (0.1, 1, 10, 50 and 100 nM). Lipid accumulation was quantified spectrophotometrically (excitation: 544 nm, emission: 590 nm). C/EBPβ, PPAR-γ1, CD36, CPT-1A, and ABCA1 protein levels were assessed by ELISA kits at different time-points (1, 2, 4, 8, and 24 h). VD3 at 50 and 100 nM significantly reduced fatty acids accumulation in macrophages by 27% and 32%, respectively. In addition, tested at 50 nM, VD3 decreased CD36, PPAR-γ1, and C/EBPβ, while it increased ABCA1 and CPT-1A protein levels in free fatty acid-exposed cells. In conclusion, VD3 reduced fatty acid accumulation in THP-1-derived macrophages exposed to lipid excess. The anti-atherogenic effect of VD3 could be ascribable to the regulation of proteins involved in lipid transport and clearance.
Collapse
|
167
|
TKTL1 Knockdown Impairs Hypoxia-Induced Glucose-6-phosphate Dehydrogenase and Glyceraldehyde-3-phosphate Dehydrogenase Overexpression. Int J Mol Sci 2022; 23:ijms23073574. [PMID: 35408935 PMCID: PMC8999113 DOI: 10.3390/ijms23073574] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/02/2022] Open
Abstract
Increased expression of transketolase (TKT) and its isoform transketolase-like-1 (TKTL1) has been related to the malignant leukemia phenotype through promoting an increase in the non-oxidative branch of the pentose phosphate pathway (PPP). Recently, it has also been described that TKTL1 can have a role in survival under hypoxic conditions and in the acquisition of radio resistance. However, TKTL1’s role in triggering metabolic reprogramming under hypoxia in leukemia cells has never been characterized. Using THP-1 AML cells, and by combining metabolomics and transcriptomics techniques, we characterized the impact of TKTL1 knockdown on the metabolic reprogramming triggered by hypoxia. Results demonstrated that TKTL1 knockdown results in a decrease in TKT, glucose-6-phosphate dehydrogenase (G6PD) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) activities and impairs the hypoxia-induced overexpression of G6PD and GAPDH, all having significant impacts on the redox capacity of NADPH- and NADH-related cells. Moreover, TKTL1 knockdown impedes hypoxia-induced transcription of genes encoding key enzymes and transporters involved in glucose, PPP and amino acid metabolism, rendering cells unable to switch to enhanced glycolysis under hypoxia. Altogether, our results show that TKTL1 plays a key role in the metabolic adaptation to hypoxia in THP-1 AML cells through modulation of G6PD and GAPDH activities, both regulating glucose/glutamine consumption and the transcriptomic overexpression of key players of PPP, glucose and amino acids metabolism.
Collapse
|
168
|
Yu C, Zhao W, Duan C, Xie J, Yin W. Poly-l-lysine-caused cell adhesion induces pyroptosis in THP-1 monocytes. Open Life Sci 2022; 17:279-283. [PMID: 35415237 PMCID: PMC8951213 DOI: 10.1515/biol-2022-0028] [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: 04/07/2021] [Revised: 11/21/2021] [Accepted: 01/03/2022] [Indexed: 11/15/2022] Open
Abstract
Pyroptosis is a kind of cell necrosis mediated by inflammasomes. The caspase 1-induced cleavage of gasdermin D (GSDMD) is a canonical pathway to cause membrane pores and eventually cell pyroptosis. Poly-l-lysine (PLL) is widely used to enhance cell adhesion during experiments. Human THP-1 cells are a typical cell line used to study pyroptosis due to their monocytic and macrophage-like characteristics. However, it was found that THP-1 cells seeded on the PLL-coated slides died. To figure out the reason, we observed the morphology of THP-1 cells on PLL-coated slides, which showed obvious pore forming on the cell membranes and cell swelling. The indicated pyroptosis-related protein expression was evaluated and it showed that the conventional caspase-1 pathway of pyroptosis was activated through the NLRP3 inflammasome in THP-1 monocytes on the PLL-coated slides. Hence, PLL-guided cell adhesion induces cell pyroptosis in THP-1 monocytes, which calls for THP-1 dominant studies of pyroptosis to avoid the use of PLL-coated slides or PLL-related drugs.
Collapse
Affiliation(s)
- Chaoping Yu
- Department of Emergency, Xijing Hospital, Fourth Military Medical University , 127 West Changle Road , Xi’an , Shaanxi 710032 , China
| | - Wei Zhao
- Department of Emergency, Xijing Hospital, Fourth Military Medical University , 127 West Changle Road , Xi’an , Shaanxi 710032 , China
| | - Chujun Duan
- Department of Emergency, Xijing Hospital, Fourth Military Medical University , 127 West Changle Road , Xi’an , Shaanxi 710032 , China
| | - Jiangang Xie
- Department of Emergency, Xijing Hospital, Fourth Military Medical University , 127 West Changle Road , Xi’an , Shaanxi 710032 , China
| | - Wen Yin
- Department of Emergency, Xijing Hospital, Fourth Military Medical University , 127 West Changle Road , Xi’an , Shaanxi 710032 , China
| |
Collapse
|
169
|
Henry B, Klement W, Gohir W, Aguilar C, Husain S. Antifungals influence the immune-related transcriptomic landscape of human monocytes after Aspergillus fumigatus infection. Sci Rep 2022; 12:4581. [PMID: 35301416 PMCID: PMC8931103 DOI: 10.1038/s41598-022-08738-4] [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: 10/19/2021] [Accepted: 02/25/2022] [Indexed: 11/26/2022] Open
Abstract
The heterogeneity of clinical responses to antifungals in aspergillosis is partially understood. We hypothesized that besides direct antifungal effects, these discrepancies may be related to different immunomodulatory profiles. Human THP-1 monocytes were coincubated in vitro with Aspergillus fumigatus and variable concentrations of voriconazole (0.5, 1 and 2 mg/l), caspofungin (1 and 2 mg/l), amphotericin B deoxycholate (0.25, 0.5 and 1 mg/l) and liposomal amphotericin B (1, 2 and 3 mg/l). After 6 h of coincubation, total cellular RNA was extracted, converted into cDNA, and transcription of 84 genes involved in antifungal immunity was measured through RT-qPCR. The presence of A. fumigatus was the main driver of the global immune-related transcriptomic response. After Aspergillus infection, thirty genes were upregulated, while 19 genes were downregulated. Discrepancies across antifungals were also evident; voriconazole-containing conditions showed similar reaction to natural infection, while the use of liposomal Amphotericin B significantly decreased the inflammatory response. Chemokines (notably CCL20 and CXCL2) and pro-inflammatory cytokines (IL1A, IL1B, IL23, G-CSF) exhibited the most pronounced differences across antifungals. Pattern recognition receptors and adaptor protein transcription were minimally affected. Protein–protein-interaction network analysis showed that IL23A played a dominant role in upregulated genes. Pathway enrichment analysis indicated that cytokine-cytokine receptor integration, TNF signaling pathways and Toll-like receptor pathways were highly involved. This exploratory study confirms the heterogeneous immunomodulatory role of antifungals. Overall, voriconazole appears to maintain an early pro-inflammatory response seen in natural infection. Assessment of immunomodulatory response with clinical response may provide a better rationale for differences observed across antifungals.
Collapse
Affiliation(s)
- Benoît Henry
- Transplant Infectious Diseases, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, 585 University Avenue, 11 PMB 138, Toronto, ON, M5G 2N2, Canada
| | - William Klement
- Transplant Infectious Diseases, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, 585 University Avenue, 11 PMB 138, Toronto, ON, M5G 2N2, Canada.,Faculty of Computer Science, Dalhousie University, Halifax, NS, Canada
| | - Wajiha Gohir
- Transplant Infectious Diseases, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, 585 University Avenue, 11 PMB 138, Toronto, ON, M5G 2N2, Canada
| | - Claire Aguilar
- Transplant Infectious Diseases, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, 585 University Avenue, 11 PMB 138, Toronto, ON, M5G 2N2, Canada.,Service des Maladies Infectieuses et Tropicales, Centre d'Infectiologie Necker Pasteur, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, France
| | - Shahid Husain
- Transplant Infectious Diseases, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, 585 University Avenue, 11 PMB 138, Toronto, ON, M5G 2N2, Canada.
| |
Collapse
|
170
|
Desheva Y, Leontieva G, Kramskaya T, Losev I, Petkova N, Rekstin A, Suvorov A. Associated virus-bacterial vaccine based on seasonal LAIV and S. pneumoniae chimeric peptide provide protection against post-influenza pneumococcal infection in mouse model. Virulence 2022; 13:558-568. [PMID: 35266442 PMCID: PMC8920162 DOI: 10.1080/21505594.2022.2049496] [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] [Indexed: 11/01/2022] Open
Abstract
Severe influenza complications are often caused by Streptococcus pneumoniae infection, which presents the most common cause of community-acquired pneumonia. We evaluated in a mouse model an associated virus-bacterial vaccine based on seasonal live influenza vaccines (LAIV) and S. pneumoniae chimeric protein comprising flagellin (PSPF). Intranasal immunization of mice with a complex of trivalent LAIV and PSPF caused an increased release of early cytokines in the lungs of mice. The immunogenicity of LAIV and PSPF in the associated vaccine composition was sometimes decreased compared to each vaccine preparation alone. Nevertheless, only vaccination of mice with LAIV+PSPF significantly reduced lethality and the bacterial load in the lungs in a model of post-influenza bacterial pneumonia. The study of the interactions of influenza viruses with bacterial peptides is important during the development of associated virus-bacterial vaccines intended for the prevention of severe post-influenza bacterial complications.
Collapse
Affiliation(s)
- Yulia Desheva
- Virology Department, FSBSI «Institute of Experimental Medicine», Saint Petersburg, Russian Federation
| | - Galina Leontieva
- Molecular Microbiology Department, FSBSI «Institute of Experimental Medicine», Saint Petersburg, Russian Federation
| | - Tatiana Kramskaya
- Molecular Microbiology Department, FSBSI «Institute of Experimental Medicine», Saint Petersburg, Russian Federation
| | - Igor Losev
- Virology Department, FSBSI «Institute of Experimental Medicine», Saint Petersburg, Russian Federation
| | - Nadezhda Petkova
- Virology Department, FSBSI «Institute of Experimental Medicine», Saint Petersburg, Russian Federation
| | - Andrey Rekstin
- Virology Department, FSBSI «Institute of Experimental Medicine», Saint Petersburg, Russian Federation
| | - Alexander Suvorov
- Molecular Microbiology Department, FSBSI «Institute of Experimental Medicine», Saint Petersburg, Russian Federation
| |
Collapse
|
171
|
Wang M, Li W, Luo Z, Tang G, Mu X, Kuang X, Guo J, Zhao Z, Flores RS, Jiang Z, Lian L, Japo JO, Ghaemmaghami AM, Zhang YS. A multifunctional micropore-forming bioink with enhanced anti-bacterial and anti-inflammatory properties. Biofabrication 2022; 14:10.1088/1758-5090/ac5936. [PMID: 35226880 PMCID: PMC8962756 DOI: 10.1088/1758-5090/ac5936] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/28/2022] [Indexed: 12/12/2022]
Abstract
Three-dimensional (3D) bioprinting has emerged as an enabling tool for various biomedical applications, such as tissue regeneration and tissue model engineering. To this end, the development of bioinks with multiple functions plays a crucial role in the applications of 3D bioprinting technologies. In this study, we propose a new bioink based on two immiscible aqueous phases of gelatin methacryloyl (GelMA) and dextran, further endowed with anti-bacterial and anti-inflammatory properties. This micropore-forming GelMA-dextran (PGelDex) bioink exhibited excellent printability with vat-polymerization, extrusion, and handheld bioprinting methods. The porous structure was confirmed after bioprinting, which promoted the spreading of the encapsulated cells, exhibiting the exceptional cytocompatibility of this bioink formulation. To extend the applications of such a micropore-forming bioink, interleukin-4 (IL-4)-loaded silver-coated gold nanorods (AgGNRs) and human mesenchymal stem cells (MSCs) were simultaneously incorporated, to display synergistic anti-infection behavior and immunomodulatory function. The results revealed the anti-bacterial properties of the AgGNR-loaded PGelDex bioink for both Gram-negative and Gram-positive bacteria. The data also indicated that the presence of IL-4 and MSCs facilitated macrophage M2-phenotype differentiation, suggesting the potential anti-inflammatory feature of the bioink. Overall, this unique anti-bacterial and immunomodulatory micropore-forming bioink offers an effective strategy for the inhibition of bacterial-induced infections as well as the ability of immune-regulation, which is a promising candidate for broadened tissue bioprinting applications.
Collapse
Affiliation(s)
- Mian Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Wanlu Li
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Zeyu Luo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Guosheng Tang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Xuan Mu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Xiao Kuang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Jie Guo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Zhibo Zhao
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Regina Sanchez Flores
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Zewei Jiang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Liming Lian
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Julia Olga Japo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| | - Amir M Ghaemmaghami
- Immunology and Immuno-bioengineering Group, School of Life Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States of America
| |
Collapse
|
172
|
Mpekoulis G, Tsopela V, Chalari A, Kalliampakou KI, Panos G, Frakolaki E, Milona RS, Sideris DC, Vassilacopoulou D, Vassilaki N. Dengue Virus Replication Is Associated with Catecholamine Biosynthesis and Metabolism in Hepatocytes. Viruses 2022; 14:v14030564. [PMID: 35336971 PMCID: PMC8948859 DOI: 10.3390/v14030564] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 12/10/2022] Open
Abstract
Previously, the association between the catecholamine biosynthetic enzyme L-Dopa decarboxylase (DDC) and Dengue virus (DV) replication was demonstrated in liver cells and was found to be mediated at least by the interaction between DDC and phosphoinositide 3-kinase (PI3K). Here, we show that biogenic amines production and uptake impede DV replication in hepatocytes and monocytes, while the virus reduces catecholamine biosynthesis, metabolism, and transport. To examine how catecholamine biosynthesis/metabolism influences DV, first, we verified the role of DDC by altering DDC expression. DDC silencing enhanced virus replication, but not translation, attenuated the negative effect of DDC substrates on the virus and reduced the infection related cell death. Then, the role of the downstream steps of the catecholamine biosynthesis/metabolism was analyzed by chemical inhibition of the respective enzymes, application of their substrates and/or their products; moreover, reserpine, the inhibitor of the vesicular monoamine transporter 2 (VMAT2), was used to examine the role of uptake/storage of catecholamines on DV. Apart from the role of each enzyme/transporter, these studies revealed that the dopamine uptake, and not the dopamine-signaling, is responsible for the negative effect on DV. Accordingly, all treatments expected to enhance the accumulation of catecholamines in the cell cytosol suppressed DV replication. This was verified by the use of chemical inducers of catecholamine biosynthesis. Last, the cellular redox alterations due to catecholamine oxidation were not related with the inhibition of DV replication. In turn, DV apart from its negative impact on DDC, inhibits tyrosine hydroxylase, dopamine beta-hydroxylase, monoamine oxidase, and VMAT2 expression.
Collapse
Affiliation(s)
- George Mpekoulis
- Laboratory of Molecular Virology, Hellenic Pasteur Institute, 11521 Athens, Greece; (G.M.); (V.T.); (A.C.); (K.I.K.); (G.P.); (R.S.M.)
| | - Vassilina Tsopela
- Laboratory of Molecular Virology, Hellenic Pasteur Institute, 11521 Athens, Greece; (G.M.); (V.T.); (A.C.); (K.I.K.); (G.P.); (R.S.M.)
| | - Anna Chalari
- Laboratory of Molecular Virology, Hellenic Pasteur Institute, 11521 Athens, Greece; (G.M.); (V.T.); (A.C.); (K.I.K.); (G.P.); (R.S.M.)
| | - Katerina I. Kalliampakou
- Laboratory of Molecular Virology, Hellenic Pasteur Institute, 11521 Athens, Greece; (G.M.); (V.T.); (A.C.); (K.I.K.); (G.P.); (R.S.M.)
| | - Georgios Panos
- Laboratory of Molecular Virology, Hellenic Pasteur Institute, 11521 Athens, Greece; (G.M.); (V.T.); (A.C.); (K.I.K.); (G.P.); (R.S.M.)
| | - Efseveia Frakolaki
- Laboratory of Molecular Virology, Hellenic Pasteur Institute, 11521 Athens, Greece; (G.M.); (V.T.); (A.C.); (K.I.K.); (G.P.); (R.S.M.)
| | - Raphaela S. Milona
- Laboratory of Molecular Virology, Hellenic Pasteur Institute, 11521 Athens, Greece; (G.M.); (V.T.); (A.C.); (K.I.K.); (G.P.); (R.S.M.)
| | - Diamantis C. Sideris
- Section of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece; (D.C.S.); (D.V.)
| | - Dido Vassilacopoulou
- Section of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece; (D.C.S.); (D.V.)
| | - Niki Vassilaki
- Laboratory of Molecular Virology, Hellenic Pasteur Institute, 11521 Athens, Greece; (G.M.); (V.T.); (A.C.); (K.I.K.); (G.P.); (R.S.M.)
- Correspondence: ; Tel.: +30-210-647-8875
| |
Collapse
|
173
|
Abstract
Human cytomegalovirus (HCMV) is a highly prevalent beta-herpesvirus and a significant cause of morbidity and mortality following hematopoietic and solid organ transplant, as well as the leading viral cause of congenital abnormalities. A key feature of the pathogenesis of HCMV is the ability of the virus to establish a latent infection in hematopoietic progenitor and myeloid lineage cells. The study of HCMV latency has been hampered by difficulties in obtaining and culturing primary cells, as well as an inability to quantitatively measure reactivating virus, but recent advances in both in vitro and in vivo models of HCMV latency and reactivation have led to a greater understanding of the interplay between host and virus. Key differences in established model systems have also led to controversy surrounding the role of viral gene products in latency establishment, maintenance, and reactivation. This review will discuss the details and challenges of various models including hematopoietic progenitor cells, monocytes, cell lines, and humanized mice. We highlight the utility and functional differences between these models and the necessary experimental design required to define latency and reactivation, which will help to generate a more complete picture of HCMV infection of myeloid-lineage cells.
Collapse
|
174
|
Giulimondi F, Vulpis E, Digiacomo L, Giuli MV, Mancusi A, Capriotti AL, Laganà A, Cerrato A, Zenezini Chiozzi R, Nicoletti C, Amenitsch H, Cardarelli F, Masuelli L, Bei R, Screpanti I, Pozzi D, Zingoni A, Checquolo S, Caracciolo G. Opsonin-Deficient Nucleoproteic Corona Endows UnPEGylated Liposomes with Stealth Properties In Vivo. ACS NANO 2022; 16:2088-2100. [PMID: 35040637 PMCID: PMC8867903 DOI: 10.1021/acsnano.1c07687] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/14/2022] [Indexed: 05/21/2023]
Abstract
For several decades, surface grafted polyethylene glycol (PEG) has been a go-to strategy for preserving the synthetic identity of liposomes in physiological milieu and preventing clearance by immune cells. However, the limited clinical translation of PEGylated liposomes is mainly due to the protein corona formation and the subsequent modification of liposomes' synthetic identity, which affects their interactions with immune cells and blood residency. Here we exploit the electric charge of DNA to generate unPEGylated liposome/DNA complexes that, upon exposure to human plasma, gets covered with an opsonin-deficient protein corona. The final product of the synthetic process is a biomimetic nanoparticle type covered by a proteonucleotidic corona, or "proteoDNAsome", which maintains its synthetic identity in vivo and is able to slip past the immune system more efficiently than PEGylated liposomes. Accumulation of proteoDNAsomes in the spleen and the liver was lower than that of PEGylated systems. Our work highlights the importance of generating stable biomolecular coronas in the development of stealth unPEGylated particles, thus providing a connection between the biological behavior of particles in vivo and their synthetic identity.
Collapse
Affiliation(s)
- Francesca Giulimondi
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Elisabetta Vulpis
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Luca Digiacomo
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Maria Valeria Giuli
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Angelica Mancusi
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Anna Laura Capriotti
- Department
of Chemistry, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Aldo Laganà
- Department
of Chemistry, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Andrea Cerrato
- Department
of Chemistry, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Riccardo Zenezini Chiozzi
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The Netherlands
| | - Carmine Nicoletti
- Unit
of Histology and Medical Embryology, Department of Anatomy, Histology,
Forensic Medicine and Orthopedics, Sapienza
University of Rome, Viale A. Scarpa 16, 00161 Rome, Italy
| | - Heinz Amenitsch
- Institute
of inorganic Chemistry, Graz University
of Technology, Stremayerg 6/IV, 8010 Graz, Austria
| | | | - Laura Masuelli
- Department
of Experimental Medicine, University of
Rome “Sapienza”, Viale Regina Elena 324, 00161 Rome, Italy
| | - Roberto Bei
- Department
of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Isabella Screpanti
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Daniela Pozzi
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Alessandra Zingoni
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Saula Checquolo
- Department
of Medico-Surgical Sciences and Biotechnology, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy
| | - Giulio Caracciolo
- Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| |
Collapse
|
175
|
Gamain B, Brousse C, Rainey NE, Diallo BK, Paquereau CE, Desrames A, Ceputyte J, Semblat JP, Bertrand O, Gangnard S, Teillaud JL, Chêne A. BMFPs, a versatile therapeutic tool for redirecting a preexisting Epstein-Barr virus antibody response toward defined target cells. SCIENCE ADVANCES 2022; 8:eabl4363. [PMID: 35148183 PMCID: PMC8836820 DOI: 10.1126/sciadv.abl4363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/17/2021] [Indexed: 06/14/2023]
Abstract
Industrial production of therapeutic monoclonal antibodies is mostly performed in eukaryotic-based systems, allowing posttranslational modifications mandatory for their functional activity. The resulting elevated product cost limits therapy access to some patients. To address this limitation, we conceptualized a novel immunotherapeutic approach to redirect a preexisting polyclonal antibody response against Epstein-Barr virus (EBV) toward defined target cells. We engineered and expressed in bacteria bimodular fusion proteins (BMFPs) comprising an Fc-deficient binding moiety targeting an antigen expressed at the surface of a target cell, fused to the EBV-P18 antigen, which recruits circulating endogenous anti-P18 IgG in EBV+ individuals. Opsonization of BMFP-coated targets efficiently triggered antibody-mediated clearing effector mechanisms. When assessed in a P18-primed mouse tumor model, therapy performed with an anti-huCD20 BMFP significantly led to increased survival and total cancer remission in some animals. These results indicate that BMFPs could represent potent and useful therapeutic molecules to treat a number of diseases.
Collapse
Affiliation(s)
- Benoît Gamain
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Carine Brousse
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Nathan E. Rainey
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Béré K. Diallo
- Laboratory “Immune Microenvironment and Immunotherapy”, INSERM U.1135, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Faculté de Médecine, Sorbonne Université, 91 boulevard de l’Hôpital, 75013 Paris, France
| | - Clara-Eva Paquereau
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Alexandra Desrames
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Jolita Ceputyte
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Jean-Philippe Semblat
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Olivier Bertrand
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Stéphane Gangnard
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Jean-Luc Teillaud
- Laboratory “Immune Microenvironment and Immunotherapy”, INSERM U.1135, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Faculté de Médecine, Sorbonne Université, 91 boulevard de l’Hôpital, 75013 Paris, France
| | - Arnaud Chêne
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| |
Collapse
|
176
|
Kužílková D, Puñet-Ortiz J, Aui PM, Fernández J, Fišer K, Engel P, van Zelm MC, Kalina T. Standardization of Workflow and Flow Cytometry Panels for Quantitative Expression Profiling of Surface Antigens on Blood Leukocyte Subsets: An HCDM CDMaps Initiative. Front Immunol 2022; 13:827898. [PMID: 35222411 PMCID: PMC8874145 DOI: 10.3389/fimmu.2022.827898] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Background The Human Cell Differentiation Molecules (HCDM) organizes Human Leukocyte Differentiation Antigen (HLDA) workshops to test and name clusters of antibodies that react with a specific antigen. These cluster of differentiation (CD) markers have provided the scientific community with validated antibody clones, consistent naming of targets and reproducible identification of leukocyte subsets. Still, quantitative CD marker expression profiles and benchmarking of reagents at the single-cell level are currently lacking. Objective To develop a flow cytometric procedure for quantitative expression profiling of surface antigens on blood leukocyte subsets that is standardized across multiple research laboratories. Methods A high content framework to evaluate the titration and reactivity of Phycoerythrin (PE)-conjugated monoclonal antibodies (mAbs) was created. Two flow cytometry panels were designed: an innate cell tube for granulocytes, dendritic cells, monocytes, NK cells and innate lymphoid cells (12-color) and an adaptive lymphocyte tube for naive and memory B and T cells, including TCRγδ+, regulatory-T and follicular helper T cells (11-color). The potential of these 2 panels was demonstrated via expression profiling of selected CD markers detected by PE-conjugated antibodies and evaluated using 561 nm excitation. Results Using automated data annotation and dried backbone reagents, we reached a robust workflow amenable to processing hundreds of measurements in each experiment in a 96-well plate format. The immunophenotyping panels enabled discrimination of 27 leukocyte subsets and quantitative detection of the expression of PE-conjugated CD markers of interest that could quantify protein expression above 400 units of antibody binding capacity. Expression profiling of 4 selected CD markers (CD11b, CD31, CD38, CD40) showed high reproducibility across centers, as well as the capacity to benchmark unique clones directed toward the same CD3 antigen. Conclusion We optimized a procedure for quantitative expression profiling of surface antigens on blood leukocyte subsets. The workflow, bioinformatics pipeline and optimized flow panels enable the following: 1) mapping the expression patterns of HLDA-approved mAb clones to CD markers; 2) benchmarking new antibody clones to established CD markers; 3) defining new clusters of differentiation in future HLDA workshops.
Collapse
Affiliation(s)
- Daniela Kužílková
- Childhood Leukaemia Investigation Prague (CLIP), Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic and University Hospital Motol, Prague, Czechia
| | - Joan Puñet-Ortiz
- Department of Biomedical Sciences, University of Barcelona, Barcelona, Spain
| | - Pei M. Aui
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Javier Fernández
- Department of Biomedical Sciences, University of Barcelona, Barcelona, Spain
| | - Karel Fišer
- Childhood Leukaemia Investigation Prague (CLIP), Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic and University Hospital Motol, Prague, Czechia
| | - Pablo Engel
- Department of Biomedical Sciences, University of Barcelona, Barcelona, Spain
| | - Menno C. van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Monash University and Alfred Hospital, Melbourne, VIC, Australia
| | - Tomáš Kalina
- Childhood Leukaemia Investigation Prague (CLIP), Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic and University Hospital Motol, Prague, Czechia
- *Correspondence: Tomáš Kalina,
| |
Collapse
|
177
|
Kulicke CA, De Zan E, Hein Z, Gonzalez-Lopez C, Ghanwat S, Veerapen N, Besra GS, Klenerman P, Christianson JC, Springer S, Nijman SM, Cerundolo V, Salio M. The P5-type ATPase ATP13A1 modulates major histocompatibility complex I-related protein 1 (MR1)-mediated antigen presentation. J Biol Chem 2022; 298:101542. [PMID: 34968463 PMCID: PMC8808182 DOI: 10.1016/j.jbc.2021.101542] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 11/08/2022] Open
Abstract
The monomorphic antigen-presenting molecule major histocompatibility complex-I-related protein 1 (MR1) presents small-molecule metabolites to mucosal-associated invariant T (MAIT) cells. The MR1-MAIT cell axis has been implicated in a variety of infectious and noncommunicable diseases, and recent studies have begun to develop an understanding of the molecular mechanisms underlying this specialized antigen presentation pathway. However, proteins regulating MR1 folding, loading, stability, and surface expression remain to be identified. Here, we performed a gene trap screen to discover novel modulators of MR1 surface expression through insertional mutagenesis of an MR1-overexpressing clone derived from the near-haploid human cell line HAP1 (HAP1.MR1). The most significant positive regulators identified included β2-microglobulin, a known regulator of MR1 surface expression, and ATP13A1, a P5-type ATPase in the endoplasmic reticulum (ER) not previously known to be associated with MR1-mediated antigen presentation. CRISPR/Cas9-mediated knockout of ATP13A1 in both HAP1.MR1 and THP-1 cell lines revealed a profound reduction in MR1 protein levels and a concomitant functional defect specific to MR1-mediated antigen presentation. Collectively, these data are consistent with the ER-resident ATP13A1 being a key posttranscriptional determinant of MR1 surface expression.
Collapse
Affiliation(s)
- Corinna A Kulicke
- MRC Human Immunology Unit, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.
| | - Erica De Zan
- Nuffield Department of Medicine, Ludwig Institute for Cancer Research Ltd and Target Discovery Institute, University of Oxford, Oxford, United Kingdom
| | - Zeynep Hein
- Department of Life Sciences and Chemistry, Jacobs University, Bremen, Germany
| | - Claudia Gonzalez-Lopez
- MRC Human Immunology Unit, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Swapnil Ghanwat
- Department of Life Sciences and Chemistry, Jacobs University, Bremen, Germany
| | - Natacha Veerapen
- School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Gurdyal S Besra
- School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Klenerman
- Peter Medawar Building, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom; Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - John C Christianson
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom
| | - Sebastian Springer
- Department of Life Sciences and Chemistry, Jacobs University, Bremen, Germany
| | - Sebastian M Nijman
- Nuffield Department of Medicine, Ludwig Institute for Cancer Research Ltd and Target Discovery Institute, University of Oxford, Oxford, United Kingdom
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Mariolina Salio
- MRC Human Immunology Unit, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
178
|
Oda T, Nakamura R, Kasamatsu T, Gotoh N, Okuda K, Saitoh T, Handa H, Murakami H, Yamashita T. DNA-double strand breaks enhance the expression of major histocompatibility complex class II through the ATM-NF-κΒ-IRF1-CIITA pathway. Cancer Gene Ther 2022; 29:225-240. [PMID: 33619341 DOI: 10.1038/s41417-021-00302-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 01/18/2021] [Accepted: 01/28/2021] [Indexed: 01/31/2023]
Abstract
Major histocompatibility complex class II (MHC II) is important for the adaptive immune response because MHC II presents processed antigens to a cluster of differentiation 4 (CD4)-positive T-cells. Conventional doses of chemotherapeutic agents induce tumor cell death by causing DNA double-strand breaks (DSBs). However, cellular responses caused by sub-lethal doses of chemotherapeutic agents are poorly understood. In this study, using low doses of chemotherapeutic agents, we showed that DSBs enhanced the expression of MHC II on cells that originate from antigen-presenting cells (APCs). These agents induced the MHC class II transactivator (CIITA), the master regulator of MHC II, and interferon regulatory factor 1 (IRF1), a transcription factor for CIITA. Short hairpin RNA against IRF1 suppressed chemotherapeutic agent-induced CIITA expression, whereas exogenous expression of IRF1 induced CIITA. Inhibition of ataxia-telangiectasia mutated (ATM), a DSB-activated kinase, suppressed induction of IRF1, CIITA, and MHC II. Similar results were observed by inhibiting NF-κB, a downstream target of ATM. These results suggest that DSBs induce MHC II activity via the ATM-NF-κB-IRF1-CIITA pathway in cells that intrinsically present antigens. Additionally, chemotherapeutic agents induced T-cell regulatory molecules. Our findings suggest that chemotherapeutic agents enhance the antigen presentation activity of APCs for T-cell activation.
Collapse
Affiliation(s)
- Tsukasa Oda
- Laboratory of Molecular Genetics, The Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan. .,Center for Food Science and Wellness, Gunma University, Maebashi, Gunma, Japan.
| | - Ruri Nakamura
- Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan
| | - Tetsuhiro Kasamatsu
- Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan
| | - Nanami Gotoh
- Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan
| | - Keiko Okuda
- Department of Molecular Diagnostics and Therapeutics, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto, Japan
| | - Takayuki Saitoh
- Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan
| | - Hiroshi Handa
- Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Hirokazu Murakami
- Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan.,Gunma University of Health and Welfare, Maebashi, Gunma, Japan
| | - Takayuki Yamashita
- Laboratory of Molecular Genetics, The Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| |
Collapse
|
179
|
Bendtsen SK, Perez-Penco M, Hübbe ML, Martinenaite E, Orebo Holmström M, Weis-Banke SE, Grønne Dahlager Jørgensen N, Jørgensen MA, Munir Ahmad S, Jensen KM, Friese C, Lundsager MT, Johansen AZ, Carretta M, Ødum N, Met Ö, Svane IM, Madsen DH, Andersen MH. Peptide vaccination activating Galectin-3-specific T cells offers a novel means to target Galectin-3-expressing cells in the tumor microenvironment. Oncoimmunology 2022; 11:2026020. [PMID: 35111385 PMCID: PMC8802901 DOI: 10.1080/2162402x.2022.2026020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Galectin-3 (Gal3) can be expressed by many cells in the tumor microenvironment (TME), including cancer cells, cancer-associated fibroblasts, tumor-associated macrophages, and regulatory T cells (Tregs). In addition to immunosuppression, Gal3 expression has been connected to malignant cell transformation, tumor progression, and metastasis. In the present study, we found spontaneous T-cell responses against Gal3-derived peptides in PBMCs from both healthy donors and cancer patients. We isolated and expanded these Gal3-specific T cells in vitro and showed that they could directly recognize target cells that expressed Gal3. Finally, therapeutic vaccination with a long Gal3-derived peptide epitope, which induced the expansion of Gal3-specific CD8+ T cells in vivo, showed a significant tumor-growth delay in mice inoculated with EO771.LMB metastatic mammary tumor cells. This was associated with a significantly lower percentage of both Tregs and tumor-infiltrating Gal3+ cells in the non-myeloid CD45+CD11b− compartment and with an alteration of the T-cell memory populations in the spleens of Gal3-vaccinated mice. These results suggest that by activating Gal3-specific T cells by an immune-modulatory vaccination, we can target Gal3-producing cells in the TME, and thereby induce a more immune permissive TME. This indicates that Gal3 could be a novel target for therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Simone Kloch Bendtsen
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Maria Perez-Penco
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Mie Linder Hübbe
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Evelina Martinenaite
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Morten Orebo Holmström
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Stine Emilie Weis-Banke
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Nicolai Grønne Dahlager Jørgensen
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Mia Aaboe Jørgensen
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Shamaila Munir Ahmad
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Kasper Mølgaard Jensen
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Christina Friese
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Mia Thorup Lundsager
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Astrid Zedlitz Johansen
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Marco Carretta
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Niels Ødum
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Özcan Met
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Daniel Hargbøl Madsen
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy (CCIT-DK), University of Copenhagen, Copenhagen University Hospital Herlev, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
180
|
Murugesan G, Davidson L, Jannetti L, Crocker PR, Weigle B. Quantitative Proteomics of Polarised Macrophages Derived from Induced Pluripotent Stem Cells. Biomedicines 2022; 10:biomedicines10020239. [PMID: 35203449 PMCID: PMC8869710 DOI: 10.3390/biomedicines10020239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
Macrophages (MΦ) are highly heterogenous and versatile innate immune cells involved in homeostatic and immune responses. Activated MΦ can exist in two extreme phenotypes: pro-inflammatory (M1) MΦ and anti-inflammatory (M2) MΦ. These phenotypes can be recapitulated in vitro by using ligands of toll-like receptors (TLRs) and cytokines such as IFNγ and IL-4. In recent years, human induced pluripotent stem cells (iPSC)-derived MΦ have gained major attention, as they are functionally similar to human monocyte-derived MΦ and are receptive to genome editing. In this study, we polarised iPSC-derived MΦ to M1 or M2 and analysed their proteome and secretome profiles using quantitative proteomics. These comprehensive proteomic data sets provide new insights into functions of polarised MΦ.
Collapse
Affiliation(s)
- Gavuthami Murugesan
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK; (G.M.); (P.R.C.)
| | - Lindsay Davidson
- Human Pluripotent Stem Cell Facility, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK;
| | - Linda Jannetti
- Division of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riss, Germany;
| | - Paul R. Crocker
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK; (G.M.); (P.R.C.)
| | - Bernd Weigle
- Division of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riss, Germany;
- Correspondence:
| |
Collapse
|
181
|
In Vitro Methodologies to Study the Role of Advanced Glycation End Products (AGEs) in Neurodegeneration. Nutrients 2022; 14:nu14020363. [PMID: 35057544 PMCID: PMC8777776 DOI: 10.3390/nu14020363] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/07/2023] Open
Abstract
Advanced glycation end products (AGEs) can be present in food or be endogenously produced in biological systems. Their formation has been associated with chronic neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and amyotrophic lateral sclerosis. The implication of AGEs in neurodegeneration is related to their ability to bind to AGE-specific receptors and the ability of their precursors to induce the so-called “dicarbonyl stress”, resulting in cross-linking and protein damage. However, the mode of action underlying their role in neurodegeneration remains unclear. While some research has been carried out in observational clinical studies, further in vitro studies may help elucidate these underlying modes of action. This review presents and discusses in vitro methodologies used in research on the potential role of AGEs in neuroinflammation and neurodegeneration. The overview reveals the main concepts linking AGEs to neurodegeneration, the current findings, and the available and advisable in vitro models to study their role. Moreover, the major questions regarding the role of AGEs in neurodegenerative diseases and the challenges and discrepancies in the research field are discussed.
Collapse
|
182
|
CD74 in Apoptotic Macrophages Is Associated with Inflammation, Plaque Progression and Clinical Manifestations in Human Atherosclerotic Lesions. Metabolites 2022; 12:metabo12010054. [PMID: 35050177 PMCID: PMC8781814 DOI: 10.3390/metabo12010054] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 02/01/2023] Open
Abstract
The aim of this study was to investigate whether CD74 levels in atherosclerotic lesions are associated with inflammation, apoptosis, plaque severity, and clinical symptoms among patients with carotid atherosclerosis. We further studied whether CD74 expression is associated with apoptosis in macrophages induced by 7ketocholesterol (7keto). Sixty-one carotid samples (39 males and 22 females) were immunostained with macrophages, smooth muscle cells, CD74, ferritin, TUNEL (Terminal deoxynucleotidyl transferase dUTP nick end labeling), and thrombin receptors. Double immunocytochemistry of CD74 and caspase 3 or CD74 and Annexin V was performed on THP-1 macrophages exposed to 7keto. In human carotid plaques, CD74 expression is lesion-dependently increased and is associated with necrotic core formation and plaque rupture, clinical symptoms, macrophage apoptosis, ferritin, and thrombin receptors. CD74 levels were inversely correlated to high-density lipoproteins and statin treatment, and positively correlated to triglycerides. In THP-1 macrophages, 7keto induced a significant increase in levels of CD74, ferritin, and apoptotic cell death. This study suggests that CD74 in apoptotic macrophages is linked to inflammation and thrombosis in progression of human atherosclerotic plaques, lipid metabolism, and clinical manifestation in atherosclerosis. Surface CD74 in apoptotic macrophages and ferritin production induced by oxidized lipids may contribute to inflammation and plaque vulnerability in atherosclerosis.
Collapse
|
183
|
Transcription Factor Activation Profiles (TFAP) identify compounds promoting differentiation of Acute Myeloid Leukemia cell lines. Cell Death Dis 2022; 8:16. [PMID: 35013135 PMCID: PMC8748454 DOI: 10.1038/s41420-021-00811-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/22/2021] [Accepted: 12/13/2021] [Indexed: 11/26/2022]
Abstract
Repurposing of drugs for new therapeutic use has received considerable attention for its potential to limit time and cost of drug development. Here we present a new strategy to identify chemicals that are likely to promote a desired phenotype. We used data from the Connectivity Map (CMap) to produce a ranked list of drugs according to their potential to activate transcription factors that mediate myeloid differentiation of leukemic progenitor cells. To validate our strategy, we tested the in vitro differentiation potential of candidate compounds using the HL-60 human cell line as a myeloid differentiation model. Ten out of 22 compounds, which were ranked high in the inferred list, were confirmed to promote significant differentiation of HL-60. These compounds may be considered candidate for differentiation therapy. The method that we have developed is versatile and it can be adapted to different drug repurposing projects.
Collapse
|
184
|
Zwicker P, Schmidt T, Hornschuh M, Lode H, Kramer A, Müller G. In vitro response of THP-1 derived macrophages to antimicrobially effective PHMB-coated Ti6Al4V alloy implant material with and without contamination with S. epidermidis and P. aeruginosa. Biomater Res 2022; 26:1. [PMID: 35000621 PMCID: PMC8744236 DOI: 10.1186/s40824-021-00247-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 12/05/2021] [Indexed: 12/23/2022] Open
Abstract
AIM Periprosthetic joint infections are a devastating complication after arthroplasty, leading to rejection of the prosthesis. The prevention of septic loosening may be possible by an antimicrobial coating of the implant surface. Poly (hexamethylene) biguanide hydrochloride [PHMB] seems to be a suitable antiseptic agent for this purpose since previous studies revealed a low cytotoxicity and a long-lasting microbicidal effect of Ti6Al4V alloy coated with PHMB. To preclude an excessive activation of the immune system, possible inflammatory effects on macrophages upon contact with PHMB-coated surfaces alone and after killing of S. epidermidis and P. aeruginosa are analyzed. METHODS THP-1 monocytes were differentiated to M0 macrophages by phorbol 12-myristate 13-acetate and seeded onto Ti6Al4V surfaces coated with various amounts of PHMB. Next to microscopic immunofluorescence analysis of labeled macrophages after adhesion on the coated surface, measurement of intracellular reactive oxygen species and analysis of cytokine secretion at different time points without and with previous bacterial contamination were conducted. RESULTS No influence on morphology of macrophages and only slight increases in iROS generation were detected. The cytokine secretion pattern depends on the surface treatment procedure and the amount of adsorbed PHMB. The PHMB coating resulted in a high reduction of viable bacteria, resulting in no significant differences in cytokine secretion as reaction to coated surfaces with and without bacterial burden. CONCLUSION Ti6Al4V specimens after alkaline treatment followed by coating with 5-7 μg PHMB and specimens treated with H2O2 before PHMB-coating (4 μg) had the smallest influence on the macrophage phienotype and thus are considered as the surface with the best cytocompatibility to macrophages tested in the present study.
Collapse
Affiliation(s)
- Paula Zwicker
- Institute of Hygiene and Environmental Medicine, Ferdinand-Sauerbruch-Str., University Medicine, D-17475, Greifswald, Germany.
| | - Thomas Schmidt
- Institute of Hygiene and Environmental Medicine, Ferdinand-Sauerbruch-Str., University Medicine, D-17475, Greifswald, Germany
| | - Melanie Hornschuh
- Institute of Hygiene and Environmental Medicine, Ferdinand-Sauerbruch-Str., University Medicine, D-17475, Greifswald, Germany
| | - Holger Lode
- Department of Pediatric Hematology and Oncology, Ferdinand-Sauerbruch-Str., University Medicine, D-17475, Greifswald, Germany
| | - Axel Kramer
- Institute of Hygiene and Environmental Medicine, Ferdinand-Sauerbruch-Str., University Medicine, D-17475, Greifswald, Germany
| | - Gerald Müller
- Institute of Hygiene and Environmental Medicine, Ferdinand-Sauerbruch-Str., University Medicine, D-17475, Greifswald, Germany
| |
Collapse
|
185
|
Kassa MW, Hasang W, Rogerson SJ. Antibody-Dependent THP-1 Cell-Mediated Phagocytosis of Plasmodium falciparum-Infected Erythrocytes. Methods Mol Biol 2022; 2470:617-628. [PMID: 35881378 DOI: 10.1007/978-1-0716-2189-9_46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Antibodies that recognize variant surface antigens (VSAs) expressed on Plasmodium falciparum-infected erythrocytes (IEs) opsonize IEs for phagocytic clearance. The anti-VSA antibodies promote antibody-dependent cellular phagocytosis (ADCP) of IEs by interacting with innate immune cells. ADCP is an important immune effector mechanism of parasite clearance. ADCP can be a tool to assess the efficacy of vaccine-induced antibodies, in addition to measuring the neutralizing ability of antibodies. Here, we developed and validated an efficient and high-throughput plate-based flow cytometric assay to measure ADCP of IEs using the human monocytic THP-1 cell line. This flow cytometric assay can be used to analyze the level of naturally acquired or vaccine-induced opsonic antibodies in large cohorts.
Collapse
Affiliation(s)
- Meseret Workineh Kassa
- Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Wina Hasang
- Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Stephen John Rogerson
- Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
186
|
Gul L, Modos D, Fonseca S, Madgwick M, Thomas JP, Sudhakar P, Booth C, Stentz R, Carding SR, Korcsmaros T. Extracellular vesicles produced by the human commensal gut bacterium Bacteroides thetaiotaomicron affect host immune pathways in a cell-type specific manner that are altered in inflammatory bowel disease. J Extracell Vesicles 2022; 11:e12189. [PMID: 35064769 PMCID: PMC8783345 DOI: 10.1002/jev2.12189] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 10/04/2021] [Accepted: 12/31/2021] [Indexed: 12/14/2022] Open
Abstract
The gastrointestinal (GI) tract harbours a complex microbial community, which contributes to its homeostasis. A disrupted microbiome can cause GI-related diseases, including inflammatory bowel disease (IBD), therefore identifying host-microbe interactions is crucial for better understanding gut health. Bacterial extracellular vesicles (BEVs), released into the gut lumen, can cross the mucus layer and access underlying immune cells. To study BEV-host interactions, we examined the influence of BEVs generated by the gut commensal bacterium, Bacteroides thetaiotaomicron, on host immune cells. Single-cell RNA sequencing data and host-microbe protein-protein interaction networks were used to predict the effect of BEVs on dendritic cells, macrophages and monocytes focusing on the Toll-like receptor (TLR) pathway. We identified biological processes affected in each immune cell type and cell-type specific processes including myeloid cell differentiation. TLR pathway analysis highlighted that BEV targets differ among cells and between the same cells in healthy versus disease (ulcerative colitis) conditions. The in silico findings were validated in BEV-monocyte co-cultures demonstrating the requirement for TLR4 and Toll-interleukin-1 receptor domain-containing adaptor protein (TIRAP) in BEV-elicited NF-kB activation. This study demonstrates that both cell-type and health status influence BEV-host communication. The results and the pipeline could facilitate BEV-based therapies for the treatment of IBD.
Collapse
Affiliation(s)
| | - Dezso Modos
- Earlham Institute, NorwichNorwichUK
- Gut Microbes and Health Research ProgrammeQuadram Institute BioscienceNorwichUK
| | - Sonia Fonseca
- Gut Microbes and Health Research ProgrammeQuadram Institute BioscienceNorwichUK
| | - Matthew Madgwick
- Earlham Institute, NorwichNorwichUK
- Gut Microbes and Health Research ProgrammeQuadram Institute BioscienceNorwichUK
| | - John P. Thomas
- Earlham Institute, NorwichNorwichUK
- Department of GastroenterologyNorfolk and Norwich University HospitalNorwichUK
| | - Padhmanand Sudhakar
- Earlham Institute, NorwichNorwichUK
- Gut Microbes and Health Research ProgrammeQuadram Institute BioscienceNorwichUK
- KU Leuven Department of Chronic DiseasesMetabolism and AgeingTranslational Research Centre for Gastrointestinal Disorders (TARGID)LeuvenBelgium
| | | | - Régis Stentz
- Gut Microbes and Health Research ProgrammeQuadram Institute BioscienceNorwichUK
| | - Simon R. Carding
- Gut Microbes and Health Research ProgrammeQuadram Institute BioscienceNorwichUK
- Norwich Medical SchoolUniversity of East AngliaNorwichUK
| | - Tamas Korcsmaros
- Earlham Institute, NorwichNorwichUK
- Gut Microbes and Health Research ProgrammeQuadram Institute BioscienceNorwichUK
| |
Collapse
|
187
|
Temporal Quantitative Phosphoproteomics Profiling of Interleukin-33 Signaling Network Reveals Unique Modulators of Monocyte Activation. Cells 2022; 11:cells11010138. [PMID: 35011700 PMCID: PMC8749991 DOI: 10.3390/cells11010138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022] Open
Abstract
Interleukin-33 (IL-33), a member of the IL-1 superfamily cytokines, is an endogenous danger signal and a nuclear-associated cytokine. It is one of the essential mediators of both innate and adaptive immune responses. Aberrant IL-33 signaling has been demonstrated to play a defensive role against various infectious and inflammatory diseases. Although the signaling responses mediated by IL-33 have been previously reported, the temporal signaling dynamics are yet to be explored. To this end, we applied quantitative temporal phosphoproteomics analysis to elucidate pathways and proteins induced by IL-33 in THP-1 monocytes. Employing a TMT labeling-based quantitation and titanium dioxide (TiO2)-based phosphopeptide enrichment strategy followed by mass spectrometry analysis, we identified and quantified 9448 unique phosphopeptides corresponding to 3392 proteins that showed differential regulation. Of these, 171 protein kinases, 60 phosphatases and 178 transcription factors were regulated at different phases of IL-33 signaling. In addition to the confirmed activation of canonical signaling modules including MAPK, NFκB, PI3K/AKT modules, pathway analysis of the time-dependent phosphorylation dynamics revealed enrichment of several cellular processes, including leukocyte adhesion, response to reactive oxygen species, cell cycle checkpoints, DNA damage and repair pathways. The detailed quantitative phosphoproteomic map of IL-33 signaling will serve as a potentially useful resource to study its function in the context of inflammatory and pathological conditions.
Collapse
|
188
|
Peterson‐Reynolds C, Mantis NJ. Differential ER stress as a driver of cell fate following ricin toxin exposure. FASEB Bioadv 2022; 4:60-75. [PMID: 35024573 PMCID: PMC8728110 DOI: 10.1096/fba.2021-00005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/16/2021] [Accepted: 08/30/2021] [Indexed: 11/11/2022] Open
Abstract
Inhalation of trace amounts of ricin toxin, a plant-derived ribosome-inactivating protein, results in ablation of alveolar macrophages, widespread epithelial damage, and the onset of acute respiratory distress syndrome (ARDS). While ricin's receptors are ubiquitous, certain cell types are more sensitive to ricin-induced cell death than others for reasons that remain unclear. For example, we demonstrate in side-by-side studies that macrophage-like differentiated THP-1 (dTHP-1) cells are hyper-sensitive to ricin, while lung epithelium-derived A549 cells are relatively insensitive, even though both cell types experience similar degrees of translational inhibition and p38 MAPK activation in response to ricin. Using a variety of small molecule inhibitors, we provide evidence that ER stress contributes to ricin-mediated cytotoxicity of dTHP-1 cells, but not A549 cells. On the other hand, the insensitivity of A549 cells to ricin was overcome by the addition of (TNF)-related apoptosis-inducing ligand (TRAIL; CD253), a known stimulator of extrinsic programmed cell death. These results have implications for understanding the complex pathophysiology of ricin-induced ARDS in that they demonstrate that intrinsic (e.g., ER stress) and extrinsic (e.g., TRAIL) factors may ultimately determine the fate of specific cell types following ricin intoxication.
Collapse
Affiliation(s)
- Claire Peterson‐Reynolds
- Division of Infectious DiseasesWadsworth CenterNew York State Department of HealthAlbanyNew YorkUSA
| | - Nicholas J. Mantis
- Division of Infectious DiseasesWadsworth CenterNew York State Department of HealthAlbanyNew YorkUSA
| |
Collapse
|
189
|
Cardoso MS, Santos RF, Almeida S, Sá M, Pérez-Cabezas B, Oliveira L, Tavares J, Carmo AM. Physical Interactions With Bacteria and Protozoan Parasites Establish the Scavenger Receptor SSC4D as a Broad-Spectrum Pattern Recognition Receptor. Front Immunol 2021; 12:760770. [PMID: 35003072 PMCID: PMC8739261 DOI: 10.3389/fimmu.2021.760770] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/29/2021] [Indexed: 12/19/2022] Open
Abstract
Since the pioneering discoveries, by the Nobel laureates Jules Hoffmann and Bruce Beutler, that Toll and Toll-like receptors can sense pathogenic microorganisms and initiate, in vertebrates and invertebrates, innate immune responses against microbial infections, many other families of pattern recognition receptors (PRRs) have been described. One of such receptor clusters is composed by, if not all, at least several members of the scavenger receptor cysteine-rich (SRCR) superfamily. Many SRCR proteins are plasma membrane receptors of immune cells; however, a small subset consists of secreted receptors that are therefore in circulation. We here describe the first characterization of biological and functional roles of the circulating human protein SSC4D, one of the least scrutinized members of the family. Within leukocyte populations, SSC4D was found to be expressed by monocytes/macrophages, neutrophils, and B cells, but its production was particularly evident in epithelial cells of several organs and tissues, namely, in the kidney, thyroid, lung, placenta, intestinal tract, and liver. Similar to other SRCR proteins, SSC4D shows the capacity of physically binding to different species of bacteria, and this opsonization can increase the phagocytic capacity of monocytes. Importantly, we have uncovered the capacity of SSC4D of binding to several protozoan parasites, a singular feature seldom described for PRRs in general and here demonstrated for the first time for an SRCR family member. Overall, our study is pioneer in assigning a PRR role to SSC4D.
Collapse
Affiliation(s)
- Marcos S. Cardoso
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Rita F. Santos
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Sarah Almeida
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
- Departamento de Biologia, Universidade de Aveiro, Aveiro, Portugal
| | - Mónica Sá
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
- Doutoramento em Ciências Farmacêuticas (especialidade Microbiologia), Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Begoña Pérez-Cabezas
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Liliana Oliveira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Joana Tavares
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Alexandre M. Carmo
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
| |
Collapse
|
190
|
Investigating Pathogenicity and Virulence of Staphylococcus pettenkoferi: An Emerging Pathogen. Int J Mol Sci 2021; 22:ijms222413614. [PMID: 34948410 PMCID: PMC8704685 DOI: 10.3390/ijms222413614] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 01/22/2023] Open
Abstract
Staphylococcus pettenkoferi is a coagulase-negative Staphylococcus identified in 2002 that has been implicated in human diseases as an opportunistic pathogenic bacterium. Its multiresistant character is becoming a major health problem, yet the pathogenicity of S. pettenkoferi is poorly characterized. In this study, the pathogenicity of a S. pettenkoferi clinical isolate from diabetic foot osteomyelitis was compared with a Staphylococcus aureus strain in various in vitro and in vivo experiments. Growth kinetics were compared against S. aureus, and bacteria survival was assessed in the RAW 264.7 murine macrophage cell line, the THP-1 human leukemia monocytic cell line, and the HaCaT human keratinocyte cell line. Ex vivo analysis was performed in whole blood survival assays and in vivo assays via the infection model of zebrafish embryos. Moreover, whole-genome analysis was performed. Our results show that S. pettenkoferi was able to survive in human blood, human keratinocytes, murine macrophages, and human macrophages. S. pettenkoferi demonstrated its virulence by causing substantial embryo mortality in the zebrafish model. Genomic analysis revealed virulence factors such as biofilm-encoding genes (e.g., icaABCD; rsbUVW) and regulator-encoding genes (e.g., agr, mgrA, sarA, saeS) well characterized in S. aureus. This study thus advances the knowledge of this under-investigated pathogen and validates the zebrafish infection model for this bacterium.
Collapse
|
191
|
Correlating biological activity to thermo-structural analysis of the interaction of CTX with synthetic models of macrophage membranes. Sci Rep 2021; 11:23712. [PMID: 34887428 PMCID: PMC8660830 DOI: 10.1038/s41598-021-02552-0] [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: 07/31/2021] [Accepted: 11/11/2021] [Indexed: 11/10/2022] Open
Abstract
The important pharmacological actions of Crotoxin (CTX) on macrophages, the main toxin in the venom of Crotalus durissus terrificus, and its important participation in the control of different pathophysiological processes, have been demonstrated. The biological activities performed by macrophages are related to signaling mediated by receptors expressed on the membrane surface of these cells or opening and closing of ion channels, generation of membrane curvature and pore formation. In the present work, the interaction of the CTX complex with the cell membrane of macrophages is studied, both using biological cells and synthetic lipid membranes to monitor structural alterations induced by the protein. Here we show that CTX can penetrate THP-1 cells and induce pores only in anionic lipid model membranes, suggesting that a possible access pathway for CTX to the cell is via lipids with anionic polar heads. Considering that the selectivity of the lipid composition varies in different tissues and organs of the human body, the thermostructural studies presented here are extremely important to open new investigations on the biological activities of CTX in different biological systems.
Collapse
|
192
|
Biological Models of the Lower Human Airways-Challenges and Special Requirements of Human 3D Barrier Models for Biomedical Research. Pharmaceutics 2021; 13:pharmaceutics13122115. [PMID: 34959396 PMCID: PMC8707984 DOI: 10.3390/pharmaceutics13122115] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 11/27/2022] Open
Abstract
In our review, we want to summarize the current status of the development of airway models and their application in biomedical research. We start with the very well characterized models composed of cell lines and end with the use of organoids. An important aspect is the function of the mucus as a component of the barrier, especially for infection research. Finally, we will explain the need for a nondestructive characterization of the barrier models using TEER measurements and live cell imaging. Here, organ-on-a-chip technology offers a great opportunity for the culture of complex airway models.
Collapse
|
193
|
Maula T, Vahvelainen N, Tossavainen H, Koivunen T, T. Pöllänen M, Johansson A, Permi P, Ihalin R. Decreased temperature increases the expression of a disordered bacterial late embryogenesis abundant (LEA) protein that enhances natural transformation. Virulence 2021; 12:1239-1257. [PMID: 33939577 PMCID: PMC8096337 DOI: 10.1080/21505594.2021.1918497] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/22/2021] [Accepted: 04/03/2021] [Indexed: 11/02/2022] Open
Abstract
Late embryogenesis abundant (LEA) proteins are important players in the management of responses to stressful conditions, such as drought, high salinity, and changes in temperature. Many LEA proteins do not have defined three-dimensional structures, so they are intrinsically disordered proteins (IDPs) and are often highly hydrophilic. Although LEA-like sequences have been identified in bacterial genomes, the functions of bacterial LEA proteins have been studied only recently. Sequence analysis of outer membrane interleukin receptor I (BilRI) from the oral pathogen Aggregatibacter actinomycetemcomitans indicated that it shared sequence similarity with group 3/3b/4 LEA proteins. Comprehensive nuclearcgq magnetic resonance (NMR) studies confirmed its IDP nature, and expression studies in A. actinomycetemcomitans harboring a red fluorescence reporter protein-encoding gene revealed that bilRI promoter expression was increased at decreased temperatures. The amino acid backbone of BilRI did not stimulate either the production of reactive oxygen species from human leukocytes or the production of interleukin-6 from human macrophages. Moreover, BilRI-specific IgG antibodies could not be detected in the sera of A. actinomycetemcomitans culture-positive periodontitis patients. Since the bilRI gene is located near genes involved in natural competence (i.e., genes associated with the uptake of extracellular (eDNA) and its incorporation into the genome), we also investigated the role of BilRI in these events. Compared to wild-type cells, the ΔbilRI mutants showed a lower transformation efficiency, which indicates either a direct or indirect role in natural competence. In conclusion, A. actinomycetemcomitans might express BilRI, especially outside the host, to survive under stressful conditions and improve its transmission potential.
Collapse
Affiliation(s)
- Terhi Maula
- Department of Life Technologies, University of Turku, Turku, Finland
| | - Nelli Vahvelainen
- Department of Life Technologies, University of Turku, Turku, Finland
| | - Helena Tossavainen
- Department of Biological and Environmental Sciences, Nanoscience Center, University of Jyvaskyla, Jyvaskyla, Finland
| | - Tuuli Koivunen
- Department of Life Technologies, University of Turku, Turku, Finland
| | | | - Anders Johansson
- Division of Molecular Periodontology, Department of Odontology, Umeå University, Umeå, Sweden
| | - Perttu Permi
- Department of Biological and Environmental Sciences, Nanoscience Center, University of Jyvaskyla, Jyvaskyla, Finland
- Department of Chemistry, Nanoscience Center, University of Jyvaskyla, Jyvaskyla, Finland
| | - Riikka Ihalin
- Department of Life Technologies, University of Turku, Turku, Finland
| |
Collapse
|
194
|
Tanner L, Mashabela GT, Omollo CC, de Wet TJ, Parkinson CJ, Warner DF, Haynes RK, Wiesner L. Intracellular Accumulation of Novel and Clinically Used TB Drugs Potentiates Intracellular Synergy. Microbiol Spectr 2021; 9:e0043421. [PMID: 34585951 PMCID: PMC8557888 DOI: 10.1128/spectrum.00434-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022] Open
Abstract
The therapeutic repertoire for tuberculosis (TB) remains limited despite the existence of many TB drugs that are highly active in in vitro models and possess clinical utility. Underlying the lack of efficacy in vivo is the inability of TB drugs to penetrate microenvironments inhabited by the causative agent, Mycobacterium tuberculosis, including host alveolar macrophages. Here, we determined the ability of the phenoxazine PhX1 previously shown to be active against M. tuberculosis in vitro to differentially penetrate murine compartments, including plasma, epithelial lining fluid, and isolated epithelial lining fluid cells. We also investigated the extent of permeation into uninfected and M. tuberculosis-infected human macrophage-like Tamm-Horsfall protein 1 (THP-1) cells directly and by comparing to results obtained in vitro in synergy assays. Our data indicate that PhX1 (4,750 ± 127.2 ng/ml) penetrates more effectively into THP-1 cells than do the clinically used anti-TB agents, rifampin (3,050 ± 62.9 ng/ml), moxifloxacin (3,374 ± 48.7 ng/ml), bedaquiline (4,410 ± 190.9 ng/ml), and linezolid (770 ± 14.1 ng/ml). Compound efficacy in infected cells correlated with intracellular accumulation, reinforcing the perceived importance of intracellular penetration as a key drug property. Moreover, we detected synergies deriving from redox-stimulatory combinations of PhX1 or clofazimine with the novel prenylated amino-artemisinin WHN296. Finally, we used compound synergies to elucidate the relationship between compound intracellular accumulation and efficacy, with PhX1/WHN296 synergy levels shown to predict drug efficacy. Collectively, our data support the utility of the applied assays in identifying in vitro active compounds with the potential for clinical development. IMPORTANCE This study addresses the development of novel therapeutic compounds for the eventual treatment of drug-resistant tuberculosis. Tuberculosis continues to progress, with cases of Mycobacterium tuberculosis (M. tuberculosis) resistance to first-line medications increasing. We assess new combinations of drugs with both oxidant and redox properties coupled with a third partner drug, with the focus here being on the potentiation of M. tuberculosis-active combinations of compounds in the intracellular macrophage environment. Thus, we determined the ability of the phenoxazine PhX1, previously shown to be active against M. tuberculosis in vitro, to differentially penetrate murine compartments, including plasma, epithelial lining fluid, and isolated epithelial lining fluid cells. In addition, the extent of permeation into human macrophage-like THP-1 cells and H37Rv-infected THP-1 cells was measured via mass spectrometry and compared to in vitro two-dimensional synergy and subsequent intracellular efficacy. Collectively, our data indicate that development of new drugs will be facilitated using the methods described herein.
Collapse
Affiliation(s)
- Lloyd Tanner
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Gabriel T. Mashabela
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Charles C. Omollo
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Timothy J. de Wet
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | | | - Digby F. Warner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Richard K. Haynes
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Lubbe Wiesner
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
195
|
Marino M, Del Bo C, Tucci M, Venturi S, Mantegazza G, Taverniti V, Møller P, Riso P, Porrini M. A mix of chlorogenic and caffeic acid reduces C/EBPß and PPAR-γ1 levels and counteracts lipid accumulation in macrophages. Eur J Nutr 2021; 61:1003-1014. [PMID: 34698900 DOI: 10.1007/s00394-021-02714-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 10/13/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Chlorogenic acid (CGA) and caffeic acid (CA) are bioactive compounds in whole grains, berries, apples, some citrus fruits and coffee, which are hypothesized to promote health-beneficial effects on the cardiovascular system. This study aimed to evaluate the capacity of CGA and CA to reduce lipid accumulation in macrophages, recognized as a critical stage in the progression of atherosclerosis. Furtherly, the modulation of CCAAT/enhancer-binding protein β (C/EBPβ) and peroxisome proliferator-activated receptor- γ1 (PPAR-γ1), as transcription factors involved in lipid metabolism, was evaluated. METHODS THP-1-derived macrophages were treated for 24 h with 0.03, 0.3, 3 and 30 μM of CGA and CA, tested alone or in combination, and a solution of oleic/palmitic acid (500 μM, 2:1 ratio). Lipid storage was assessed spectrophotometrically through fluorescent staining of cells with Nile red. C/EBPβ and PPAR-γ1 mRNA and protein levels were evaluated by RT-PCR and enzyme-linked immunosorbent assay, respectively. RESULTS The mix of CGA + CA (1:1 ratio) reduced lipid accumulation at all concentrations tested, except for the highest one. The greatest effect ( - 65%; p < 0.01) was observed at the concentration of 0.3 μM for each compound. The same concentration significantly (p < 0.01) downregulated C/EBPβ and PPAR-γ1 gene expression and reduced their protein levels at 2 h and 24 h, respectively. CONCLUSION The results indicate that the capacity of CGA + CA mix to reduce lipid storage in macrophages is mediated by a reduction in the expression of transcription factors C/EBPβ and PPAR-γ1.
Collapse
Affiliation(s)
- Mirko Marino
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università Degli Studi Di Milano, 20133, Milan, Italy
| | - Cristian Del Bo
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università Degli Studi Di Milano, 20133, Milan, Italy.
| | - Massimiliano Tucci
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università Degli Studi Di Milano, 20133, Milan, Italy
| | - Samuele Venturi
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università Degli Studi Di Milano, 20133, Milan, Italy
| | - Giacomo Mantegazza
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università Degli Studi Di Milano, 20133, Milan, Italy
| | - Valentina Taverniti
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università Degli Studi Di Milano, 20133, Milan, Italy
| | - Peter Møller
- Department of Public Health, Section of Environmental Health, University of Copenhagen, 1014, Copenhagen K, Denmark
| | - Patrizia Riso
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università Degli Studi Di Milano, 20133, Milan, Italy
| | - Marisa Porrini
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università Degli Studi Di Milano, 20133, Milan, Italy
| |
Collapse
|
196
|
Lai CY, Tseng PC, Chen CL, Satria RD, Wang YT, Lin CF. Different Induction of PD-L1 (CD274) and PD-1 (CD279) Expression in THP-1-Differentiated Types 1 and 2 Macrophages. J Inflamm Res 2021; 14:5241-5249. [PMID: 34675601 PMCID: PMC8520887 DOI: 10.2147/jir.s329921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 09/28/2021] [Indexed: 12/15/2022] Open
Abstract
Background Phorbol 12-myristate 13-acetate (PMA)-induced differentiation of human monocytic THP-1 cells is an experimental model for preparing resting macrophages (M0) for cell polarization toward the different functional specializations of macrophages. Methods In this study, we examined the expression of immune checkpoints by using flow cytometry following multicolor staining. The blockade of immune checkpoint by using neutralizing antibodies was performed to assess their role in PMA-induced THP-1-differentiated macrophages. Results Upon the inducible macrophage differentiation caused by PMA, increased expression levels of CD11b and CD68 were measured and characterized according to their adherent phenotype accompanied by the generation of cellular complexity. While the cell growth rate was abolished post-differentiation, some cells underwent cell death. Notably, we found increases in the expression of programmed cell death protein 1, also known as PD-1 (CD279), and its ligand PD-L1 (CD274), mainly in differentiated M0 (CD68+CD11b+) macrophages. However, neutralizing PD-L1/PD-1 neither blocked THP-1 cell differentiation toward macrophages nor inhibited macrophage polarization in M1 and M2. In specializing macrophages, a decrease both in CD274 and CD279 was found in M2. Conclusion These results revealed the inducible expression of PD-L1/PD-1 in PMA-induced THP-1-differentiated M0 macrophages followed by a decrease in M2 macrophages.
Collapse
Affiliation(s)
- Chun-Yi Lai
- Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei, 110, Taiwan.,Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Po-Chun Tseng
- Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei, 110, Taiwan.,Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Chia-Ling Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Rahmat Dani Satria
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Department of Clinical Pathology and Laboratory Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia.,Clinical Laboratory Installation, Dr. Sardjito Central General Hospital, Yogyakarta, 55281, Indonesia
| | - Yung-Ting Wang
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Chiou-Feng Lin
- Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei, 110, Taiwan.,Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| |
Collapse
|
197
|
Lafuente-Gracia L, Borgiani E, Nasello G, Geris L. Towards in silico Models of the Inflammatory Response in Bone Fracture Healing. Front Bioeng Biotechnol 2021; 9:703725. [PMID: 34660547 PMCID: PMC8514728 DOI: 10.3389/fbioe.2021.703725] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 09/07/2021] [Indexed: 12/21/2022] Open
Abstract
In silico modeling is a powerful strategy to investigate the biological events occurring at tissue, cellular and subcellular level during bone fracture healing. However, most current models do not consider the impact of the inflammatory response on the later stages of bone repair. Indeed, as initiator of the healing process, this early phase can alter the regenerative outcome: if the inflammatory response is too strongly down- or upregulated, the fracture can result in a non-union. This review covers the fundamental information on fracture healing, in silico modeling and experimental validation. It starts with a description of the biology of fracture healing, paying particular attention to the inflammatory phase and its cellular and subcellular components. We then discuss the current state-of-the-art regarding in silico models of the immune response in different tissues as well as the bone regeneration process at the later stages of fracture healing. Combining the aforementioned biological and computational state-of-the-art, continuous, discrete and hybrid modeling technologies are discussed in light of their suitability to capture adequately the multiscale course of the inflammatory phase and its overall role in the healing outcome. Both in the establishment of models as in their validation step, experimental data is required. Hence, this review provides an overview of the different in vitro and in vivo set-ups that can be used to quantify cell- and tissue-scale properties and provide necessary input for model credibility assessment. In conclusion, this review aims to provide hands-on guidance for scientists interested in building in silico models as an additional tool to investigate the critical role of the inflammatory phase in bone regeneration.
Collapse
Affiliation(s)
- Laura Lafuente-Gracia
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Edoardo Borgiani
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA in silico Medicine, University of Liège, Liège, Belgium
| | - Gabriele Nasello
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA in silico Medicine, University of Liège, Liège, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| |
Collapse
|
198
|
Zhen XX, Yang L, Gu Y, Yang Q, Gu WW, He YP, Wang YL, Wang J. MNSFβ Regulates TNFα Production by Interacting with RC3H1 in Human Macrophages, and Dysfunction of MNSFβ in Decidual Macrophages Is Associated With Recurrent Pregnancy Loss. Front Immunol 2021; 12:691908. [PMID: 34589082 PMCID: PMC8473736 DOI: 10.3389/fimmu.2021.691908] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/23/2021] [Indexed: 12/29/2022] Open
Abstract
Decidual macrophages (dMϕ) are the second largest population of leukocytes at the maternal–fetal interface and play critical roles in maintaining pregnancy. Our previous studies demonstrated the active involvement of monoclonal nonspecific suppressor factor-β (MNSFβ) in embryonic implantation and pregnancy success. MNSFβ is a ubiquitously expressed ubiquitin-like protein that also exhibits immune regulatory potential, but its function in human dMϕ remains unknown. Here, we observed that the proportion of CD11chigh (CD11cHI) dMϕ was significantly increased in dMϕ derived from patients with recurrent pregnancy loss (RPL dMϕ) compared to those derived from normal pregnant women (Control dMϕ). The production of MNSFβ and TNFα by RPL dMϕ was also significantly increased compared to that by Control dMϕ. Conditioned medium from RPL dMϕ exerted an inhibitory effect on the invasiveness of human trophoblastic HTR8/SVneo cells, and this effect could be partially reversed by a neutralizing antibody against TNFα. Bioinformatics analysis indicated a potential interaction between MNSFβ and RC3H1, a suppressor of TNFα transcription. Immunoprecipitation experiments with human Mϕ differentiated from the human monocyte cell line Thp1 (Thp1-derived Mϕ) proved the binding of MNSFβ to RC3H1. Specific knockdown of MNSFβ in Thp1-derived Mϕ led to a marked decrease in TNFα production, which could be reversed by inhibiting RC3H1 expression. Interestingly, a significant decrease in the protein level of RC3H1 was observed in RPL dMϕ. Together, our findings indicate that aberrantly increased MNSFβ expression in dMϕ may promote TNFα production via its interaction with RC3H1, and these phenomena could result in the disruption of the immune balance at the maternal–fetal interface and thus pregnancy loss.
Collapse
Affiliation(s)
- Xing-Xing Zhen
- National Health Commission (NHC) of the People's Republic of China Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Long Yang
- National Health Commission (NHC) of the People's Republic of China Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Yan Gu
- Department of Gynecology and Obstetrics, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Qian Yang
- National Health Commission (NHC) of the People's Republic of China Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wen-Wen Gu
- National Health Commission (NHC) of the People's Republic of China Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Ya-Ping He
- National Health Commission (NHC) of the People's Republic of China Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Yan-Ling Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Jian Wang
- National Health Commission (NHC) of the People's Republic of China Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
199
|
Wen Y, Wang X, Cahya S, Anderson P, Velasquez C, Torres C, Ferrante A, Kaliyaperumal A. Comparability study of monocyte derived dendritic cells, primary monocytes, and THP1 cells for innate immune responses. J Immunol Methods 2021; 498:113147. [PMID: 34508774 DOI: 10.1016/j.jim.2021.113147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/06/2021] [Indexed: 01/23/2023]
Abstract
Immunogenicity is one major challenge to the successful development of biotherapeutics because it could adversely affect PK/PD, safety, and efficacy. Preclinical immunogenicity risk assessment strategies and assays have been developed and implemented to screen and optimize discovery molecules. Internalization by antigen presenting cells (APC) and innate immune activation are initial prerequisite steps in eliciting immune responses to biotherapeutics. Dendritic cells (DC)- and monocyte-based assays are employed to interrogate such risks, and their value has been well documented in the literature. However, these assays have limited throughput, exhibit higher variability, and entail lengthy and complex procedures as they are based on primary cells such as peripheral blood mononuclear cells (PBMC) from individual donors. Herein, we investigated THP1 cells as surrogate cells to study APC internalization and innate immune activation. Comparability studies showed that THP1 cells could resemble innate immune responses of monocyte-derived DC and primary CD14+ monocytes using a panel of therapeutic antibodies. In addition, an automated high throughput THP1 internalization assay was qualified to enable risk assessment at pre‑lead stages. The results demonstrated that THP1 cells can be utilized to assess immunogenicity risk in a high throughput manner.
Collapse
Affiliation(s)
- Yi Wen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Xiaoli Wang
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Suntara Cahya
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Paul Anderson
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Candyd Velasquez
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Carina Torres
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Andrea Ferrante
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | | |
Collapse
|
200
|
Heduschke A, Ackermann K, Wilhelm B, Mey L, Bonaterra GA, Kinscherf R, Schwarz A. GDF-15 Deficiency Reduces Autophagic Activity in Human Macrophages In Vitro and Decreases p62-Accumulation in Atherosclerotic Lesions in Mice. Cells 2021; 10:2346. [PMID: 34571994 PMCID: PMC8470202 DOI: 10.3390/cells10092346] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/31/2021] [Accepted: 09/04/2021] [Indexed: 12/26/2022] Open
Abstract
(1) Background: Growth differentiation factor-15 (GDF-15) is associated with cardiovascular diseases and autophagy in human macrophages (MΦ). Thus, we are interested in investigating autophagic mechanisms with special respect to the role of GDF-15. (2) Methods: Recombinant (r)GDF-15 and siRNA GDF-15 were used to investigate the effects of GDF-15 on autophagic and lysosomal activity, as well as autophagosome formation by transmission electron microscopy (TEM) in MΦ. To ascertain the effects of GDF-15-/- on the progression of atherosclerotic lesions, we used GDF-15-/-/ApoE-/- and ApoE-/- mice under a cholesterol-enriched diet (CED). Body weight, body mass index (BMI), blood lipid levels and lumen stenosis in the brachiocephalic trunk (BT) were analyzed. Identification of different cell types and localization of autophagy-relevant proteins in atherosclerotic plaques were performed by immunofluorescence. (3) Results: siGDF-15 reduced and, conversely, rGDF-15 increased the autophagic activity in MΦ, whereas lysosomal activity was unaffected. Autophagic degradation after starvation and rGDF-15 treatment was observed by TEM. GDF-15-/-/ApoE-/- mice, after CED, showed reduced lumen stenosis in the BT, while body weight, BMI and triglycerides were increased compared with ApoE-/- mice. GDF-15-/- decreased p62-accumulation in atherosclerotic lesions, especially in endothelial cells (ECs). (4) Conclusion: GDF-15 seems to be an important factor in the regulation of autophagy, especially in ECs of atherosclerotic lesions, indicating its crucial pathophysiological function during atherosclerosis development.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anja Schwarz
- Institute for Anatomy and Cell Biology, Department of Medical Cell Biology, Philipps-University of Marburg, 35032 Marburg, Germany; (A.H.); (K.A.); (B.W.); (L.M.); (G.A.B.); (R.K.)
| |
Collapse
|