1
|
Wu J, Ai T, He P, Shi Q, Li Y, Zhang Z, Chen M, Huang Z, Wu S, Chen W, Han J. Cecal necroptosis triggers lethal cardiac dysfunction in TNF-induced severe SIRS. Cell Rep 2024; 43:114778. [PMID: 39325617 DOI: 10.1016/j.celrep.2024.114778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Tumor necrosis factor (TNF) induces systemic inflammatory response syndrome (SIRS), and severe SIRS can serve as a model for studying animal death caused by organ failure. Through strategic cecectomy, we demonstrate that necroptosis in the cecum initiates the death process in TNF-treated mice, but it is not the direct cause of death. Instead, we show that it is the cardiac dysfunction downstream of cecum damage that ultimately leads to the death of TNF-treated mice. By in vivo and ex vivo physiological analyses, we reveal that TNF and the damage-associated molecular patterns (DAMPs) released from necroptotic cecal cells jointly target cardiac endothelial cells, triggering caspase-8 activation and subsequent cardiac endothelial damage. Cardiac endothelial damage is a primary cause of the deterioration of diastolic function in the heart of TNF-treated mice. Our research provides insights into the pathophysiological process of TNF-induced lethality.
Collapse
Affiliation(s)
- Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China
| | - Tingting Ai
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Peng He
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Research Unit of Cellular Stress of Chinese Academy of Medical Sciences, Xiang'an Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Qilin Shi
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yangxin Li
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Ziguan Zhang
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Minwei Chen
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Zhengrong Huang
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Suqin Wu
- Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China
| | - Wanze Chen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518000, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China; Research Unit of Cellular Stress of Chinese Academy of Medical Sciences, Xiang'an Hospital of Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
2
|
Kulbay M, Paimboeuf A, Ozdemir D, Bernier J. Review of cancer cell resistance mechanisms to apoptosis and actual targeted therapies. J Cell Biochem 2021; 123:1736-1761. [PMID: 34791699 DOI: 10.1002/jcb.30173] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/04/2021] [Accepted: 10/22/2021] [Indexed: 11/11/2022]
Abstract
The apoptosis pathway is a programmed cell death mechanism that is crucial for cellular and tissue homeostasis and organ development. There are three major caspase-dependent pathways of apoptosis that ultimately lead to DNA fragmentation. Cancerous cells are known to highly regulate the apoptotic pathway and its role in cancer hallmark acquisition has been discussed over the past decades. Numerous mutations in cancer cell types have been reported to be implicated in chemoresistance and treatment outcome. In this review, we summarize the mutations of the caspase-dependant apoptotic pathways that are the source of cancer development and the targeted therapies currently available or in trial.
Collapse
Affiliation(s)
- Merve Kulbay
- INRS - Centre Armand-Frappier Santé Biotechnologie, Laval, Quebec, Canada.,Department of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Adeline Paimboeuf
- INRS - Centre Armand-Frappier Santé Biotechnologie, Laval, Quebec, Canada
| | - Derman Ozdemir
- Department of Medicine, One Brooklyn Health-Brookdale Hospital Medical Center, Brooklyn, New York, USA
| | - Jacques Bernier
- INRS - Centre Armand-Frappier Santé Biotechnologie, Laval, Quebec, Canada
| |
Collapse
|
3
|
Nelke J, Medler J, Weisenberger D, Beilhack A, Wajant H. CD40- and CD95-specific antibody single chain-Baff fusion proteins display BaffR-, TACI- and BCMA-restricted agonism. MAbs 2021; 12:1807721. [PMID: 32840410 PMCID: PMC7531569 DOI: 10.1080/19420862.2020.1807721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Antibodies that target a clinically relevant group of receptors within the tumor necrosis factor receptor superfamily (TNFRSF), including CD40 and CD95 (Fas/Apo-1), also require binding to Fc gamma receptors (FcγRs) to elicit a strong agonistic activity. This FcγR dependency largely relies on the mere cellular anchoring through the antibody’s Fc domain and does not involve the engagement of FcγR signaling. The aim of this study was to elicit agonistic activity from αCD40 and αCD95 antibodies in a myeloma cell anchoring-controlled FcγR-independent manner. For this purpose, various antibody variants (IgG1, IgG1N297A, Fab2) against the TNFRSF members CD40 and CD95 were genetically fused to a single-chain-encoded B-cell activating factor (scBaff) trimer as a C-terminal myeloma-specific anchoring domain substituting for Fc domain-mediated FcγR binding. The antibody-scBaff fusion proteins were evaluated in binding studies and functional assays using tumor cell lines expressing one or more of the three receptors of Baff: BaffR, transmembrane activator and CAML interactor (TACI) and B-cell maturation antigen (BCMA). Cellular binding studies showed that the binding properties of the different domains within the fusion proteins remained fully intact in the antibody-scBaff fusion proteins. In co-culture assays of CD40- and CD95-responsive cells with BaffR, BCMA or TACI expressing anchoring cells, the antibody fusion proteins displayed strong agonism while only minor receptor stimulation was observed in co-cultures with cells without expression of Baff-interacting receptors. Thus, our CD40 and CD95 antibody fusion proteins display myeloma cell-dependent activity and promise reduced systemic side effects compared to conventional CD40 and CD95 agonists.
Collapse
Affiliation(s)
- Johannes Nelke
- Molecular Internal Medicine, University Hospital Würzburg , Würzburg, Germany
| | - Juliane Medler
- Molecular Internal Medicine, University Hospital Würzburg , Würzburg, Germany
| | | | - Andreas Beilhack
- Interdisciplinary Center for Clinical Research, University Hospital Würzburg , Würzburg, Germany
| | - Harald Wajant
- Molecular Internal Medicine, University Hospital Würzburg , Würzburg, Germany
| |
Collapse
|
4
|
Kucka K, Wajant H. Receptor Oligomerization and Its Relevance for Signaling by Receptors of the Tumor Necrosis Factor Receptor Superfamily. Front Cell Dev Biol 2021; 8:615141. [PMID: 33644033 PMCID: PMC7905041 DOI: 10.3389/fcell.2020.615141] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/28/2020] [Indexed: 12/20/2022] Open
Abstract
With the exception of a few signaling incompetent decoy receptors, the receptors of the tumor necrosis factor receptor superfamily (TNFRSF) are signaling competent and engage in signaling pathways resulting in inflammation, proliferation, differentiation, and cell migration and also in cell death induction. TNFRSF receptors (TNFRs) become activated by ligands of the TNF superfamily (TNFSF). TNFSF ligands (TNFLs) occur as trimeric type II transmembrane proteins but often also as soluble ligand trimers released from the membrane-bound form by proteolysis. The signaling competent TNFRs are efficiently activated by the membrane-bound TNFLs. The latter recruit three TNFR molecules, but there is growing evidence that this is not sufficient to trigger all aspects of TNFR signaling; rather, the formed trimeric TNFL–TNFR complexes have to cluster secondarily in the cell-to-cell contact zone for full TNFR activation. With respect to their response to soluble ligand trimers, the signaling competent TNFRs can be subdivided into two groups. TNFRs of one group, designated as category I TNFRs, are robustly activated by soluble ligand trimers. The receptors of a second group (category II TNFRs), however, failed to become properly activated by soluble ligand trimers despite high affinity binding. The limited responsiveness of category II TNFRs to soluble TNFLs can be overcome by physical linkage of two or more soluble ligand trimers or, alternatively, by anchoring the soluble ligand molecules to the cell surface or extracellular matrix. This suggests that category II TNFRs have a limited ability to promote clustering of trimeric TNFL–TNFR complexes outside the context of cell–cell contacts. In this review, we will focus on three aspects on the relevance of receptor oligomerization for TNFR signaling: (i) the structural factors which promote clustering of free and liganded TNFRs, (ii) the signaling pathway specificity of the receptor oligomerization requirement, and (iii) the consequences for the design and development of TNFR agonists.
Collapse
Affiliation(s)
- Kirstin Kucka
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
5
|
Datta-Mannan A, Brown RM, Fitchett J, Heng AR, Balasubramaniam D, Pereira J, Croy JE. Modulation of the Biophysical Properties of Bifunctional Antibodies as a Strategy for Mitigating Poor Pharmacokinetics. Biochemistry 2019; 58:3116-3132. [DOI: 10.1021/acs.biochem.9b00074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
| | | | - Jonathan Fitchett
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, California 92121, United States
| | - Aik Roy Heng
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, California 92121, United States
| | - Deepa Balasubramaniam
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology Center, San Diego, California 92121, United States
| | | | | |
Collapse
|
6
|
Medler J, Nelke J, Weisenberger D, Steinfatt T, Rothaug M, Berr S, Hünig T, Beilhack A, Wajant H. TNFRSF receptor-specific antibody fusion proteins with targeting controlled FcγR-independent agonistic activity. Cell Death Dis 2019; 10:224. [PMID: 30833543 PMCID: PMC6399339 DOI: 10.1038/s41419-019-1456-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/10/2019] [Accepted: 02/12/2019] [Indexed: 01/08/2023]
Abstract
Antibodies specific for TNFRSF receptors that bind soluble ligands without getting properly activated generally act as strong agonists upon FcγR binding. Systematic analyses revealed that the FcγR dependency of such antibodies to act as potent agonists is largely independent from isotype, FcγR type, and of the epitope recognized. This suggests that the sole cellular attachment, achieved by Fc domain-FcγR interaction, dominantly determines the agonistic activity of antibodies recognizing TNFRSF receptors poorly responsive to soluble ligands. In accordance with this hypothesis, we demonstrated that antibody fusion proteins harboring domains allowing FcγR-independent cell surface anchoring also act as strong agonist provided they have access to their target. This finding defines a general possibility to generate anti-TNFRSF receptor antibodies with FcγR-independent agonism. Moreover, anti-TNFRSF receptor antibody fusion proteins with an anchoring domain promise superior applicability to conventional systemically active agonists when an anchoring target with localized disease associated expression can be addressed.
Collapse
Affiliation(s)
- Juliane Medler
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Auvera Haus Grombühlstraße 12, 97080, Würzburg, Germany
| | - Johannes Nelke
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Auvera Haus Grombühlstraße 12, 97080, Würzburg, Germany
| | - Daniela Weisenberger
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Auvera Haus Grombühlstraße 12, 97080, Würzburg, Germany
| | - Tim Steinfatt
- Department of Internal Medicine II, University Hospital of Würzburg, Zinklesweg 10, 97078, Würzburg, Germany
| | - Moritz Rothaug
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Auvera Haus Grombühlstraße 12, 97080, Würzburg, Germany
| | - Susanne Berr
- Institute of Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, 97078, Würzburg, Germany
| | - Thomas Hünig
- Institute of Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, 97078, Würzburg, Germany
| | - Andreas Beilhack
- Department of Internal Medicine II, University Hospital of Würzburg, Zinklesweg 10, 97078, Würzburg, Germany
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Auvera Haus Grombühlstraße 12, 97080, Würzburg, Germany.
| |
Collapse
|
7
|
Cubero FJ, Woitok MM, Zoubek ME, de Bruin A, Hatting M, Trautwein C. Disruption of the FasL/Fas axis protects against inflammation-derived tumorigenesis in chronic liver disease. Cell Death Dis 2019; 10:115. [PMID: 30737368 PMCID: PMC6368573 DOI: 10.1038/s41419-019-1391-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/16/2019] [Accepted: 01/21/2019] [Indexed: 12/21/2022]
Abstract
Fas Ligand (FasL) and Fas (APO-1/CD95) are members of the TNFR superfamily and may trigger apoptosis. Here, we aimed to elucidate the functional role of Fas signaling in an experimental model of chronic liver disease, the hepatocyte-specific NEMO knockout (NEMOΔhepa) mice. We generated NEMOΔhepa /Faslpr mice, while NEMOΔhepa, NEMOf/f as well as Faslpranimals were used as controls, and characterized their phenotype during liver disease progression. Liver damage was evaluated by serum transaminases, histological, immunofluorescence procedures, and biochemical and molecular biology techniques. Proteins were detected by western Blot, expression of mRNA by RT-PCR, and infiltration of inflammatory cells was determined by FACs analysis, respectively. Faslpr mutation in NEMOΔhepa mice resulted in overall decreased liver injury, enhanced hepatocyte survival, and reduced proliferation at 8 weeks of age compared with NEMOΔhepa mice. Moreover, NEMOΔhepa/Faslpr animals elicited significantly decreased parameters of liver fibrosis, such as Collagen IA1, MMP2, and TIMP1, and reduced proinflammatory macrophages and cytokine expression. At 52 weeks of age, NEMOΔhepa/Faslpr exhibited less malignant growth as evidenced by reduced HCC burden associated with a significantly decreased number of nodules and LW/BW ratio and decreased myeloid populations. Deletion of TNFR1 further reduced tumor load of 52-weeks-old NEMOΔhepa/Faslpr mice. The functionality of FasL/Fas might affect inflammation-driven tumorigenesis in an experimental model of chronic liver disease. These results help to develop alternative therapeutic approaches and extend the limitations of tumor therapy against HCC.
Collapse
Affiliation(s)
- Francisco Javier Cubero
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany. .,Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid, Spain. .,12 de Octubre Health Research Institute (imas12), Madrid, Spain.
| | | | - Miguel E Zoubek
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany.,Department of Toxicology, Faculty of Health Medicine and Life Sciences, School of Nutrition, Toxicology and Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Alain de Bruin
- Department of Toxicology, Faculty of Health Medicine and Life Sciences, School of Nutrition, Toxicology and Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands.,Institute of Pathology, Utrecht University, Utrecht, The Netherlands
| | - Maximilian Hatting
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany.
| |
Collapse
|
8
|
Datta-Mannan A, Croy JE, Schirtzinger L, Torgerson S, Breyer M, Wroblewski VJ. Aberrant bispecific antibody pharmacokinetics linked to liver sinusoidal endothelium clearance mechanism in cynomolgus monkeys. MAbs 2017; 8:969-82. [PMID: 27111637 DOI: 10.1080/19420862.2016.1178435] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Bispecific antibodies (BsAbs) can affect multiple disease pathways, thus these types of constructs potentially provide promising approaches to improve efficacy in complex disease indications. The specific and non-specific clearance mechanisms/biology that affect monoclonal antibody (mAb) pharmacokinetics are likely involved in the disposition of BsAbs. Despite these similarities, there are a paucity of studies on the in vivo biology that influences the biodistribution and pharmacokinetics of BsAbs. The present case study evaluated the in vivo disposition of 2 IgG-fusion BsAb formats deemed IgG-ECD (extracellular domain) and IgG-scFv (single-chain Fv) in cynomolgus monkeys. These BsAb molecules displayed inferior in vivo pharmacokinetic properties, including a rapid clearance (> 0.5 mL/hr/kg) and short half-life relative to their mAb counterparts. The current work evaluated factors in vivo that result in the aberrant clearance of these BsAb constructs. Results showed the rapid clearance of the BsAbs that was not attributable to target binding, reduced neonatal Fc receptor (FcRn) interactions or poor molecular/biochemical properties. Evaluation of the cellular distribution of the constructs suggested that the major clearance mechanism was linked to binding/association with liver sinusoidal endothelial cells (LSECs) versus liver macrophages. The role of LSECs in facilitating the clearance of the IgG-ECD and IgG-scFv BsAb constructs described in these studies was consistent with the minimal influence of clodronate-mediated macrophage depletion on the pharmacokinetics of the constructs in cynomolgus monkeys The findings in this report are an important demonstration that the elucidation of clearance mechanisms for some IgG-ECD and IgG-scFv BsAb molecules can be unique and complicated, and may require increased attention due to the proliferation of these more complex mAb-like structures.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- a Department of Drug Disposition, Development/ Commercialization , Lilly Research Laboratories, Lilly Corporate Center , Indianapolis , Indiana , USA
| | - Johnny E Croy
- b Department of Biotechnology Discovery Research , Lilly Research Laboratories, Lilly Corporate Center , Indianapolis , Indiana , USA
| | - Linda Schirtzinger
- a Department of Drug Disposition, Development/ Commercialization , Lilly Research Laboratories, Lilly Corporate Center , Indianapolis , Indiana , USA
| | - Stacy Torgerson
- a Department of Drug Disposition, Development/ Commercialization , Lilly Research Laboratories, Lilly Corporate Center , Indianapolis , Indiana , USA
| | - Matthew Breyer
- b Department of Biotechnology Discovery Research , Lilly Research Laboratories, Lilly Corporate Center , Indianapolis , Indiana , USA
| | - Victor J Wroblewski
- a Department of Drug Disposition, Development/ Commercialization , Lilly Research Laboratories, Lilly Corporate Center , Indianapolis , Indiana , USA
| |
Collapse
|
9
|
O' Reilly E, Tirincsi A, Logue SE, Szegezdi E. The Janus Face of Death Receptor Signaling during Tumor Immunoediting. Front Immunol 2016; 7:446. [PMID: 27843441 PMCID: PMC5086583 DOI: 10.3389/fimmu.2016.00446] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/07/2016] [Indexed: 12/24/2022] Open
Abstract
Cancer immune surveillance is essential for the inhibition of carcinogenesis. Malignantly transformed cells can be recognized by both the innate and adaptive immune systems through different mechanisms. Immune effector cells induce extrinsic cell death in the identified tumor cells by expressing death ligand cytokines of the tumor necrosis factor ligand family. However, some tumor cells can escape immune elimination and progress. Acquisition of resistance to the death ligand-induced apoptotic pathway can be obtained through cleavage of effector cell expressed death ligands into a poorly active form, mutations or silencing of the death receptors, or overexpression of decoy receptors and pro-survival proteins. Although the immune system is highly effective in the elimination of malignantly transformed cells, abnormal/dysfunctional death ligand signaling curbs its cytotoxicity. Moreover, DRs can also transmit pro-survival and pro-migratory signals. Consequently, dysfunctional death receptor-mediated apoptosis/necroptosis signaling does not only give a passive resistance against cell death but actively drives tumor cell motility, invasion, and contributes to consequent metastasis. This dual contribution of the death receptor signaling in both the early, elimination phase, and then in the late, escape phase of the tumor immunoediting process is discussed in this review. Death receptor agonists still hold potential for cancer therapy since they can execute the tumor-eliminating immune effector function even in the absence of activation of the immune system against the tumor. The opportunities and challenges of developing death receptor agonists into effective cancer therapeutics are also discussed.
Collapse
Affiliation(s)
- Eimear O' Reilly
- Apoptosis Research Center, School of Natural Sciences, National University of Ireland , Galway , Ireland
| | - Andrea Tirincsi
- Apoptosis Research Center, School of Natural Sciences, National University of Ireland , Galway , Ireland
| | - Susan E Logue
- Apoptosis Research Center, School of Natural Sciences, National University of Ireland , Galway , Ireland
| | - Eva Szegezdi
- Apoptosis Research Center, School of Natural Sciences, National University of Ireland , Galway , Ireland
| |
Collapse
|
10
|
Tanoi T, Tamura T, Sano N, Nakayama K, Fukunaga K, Zheng YW, Akhter A, Sakurai Y, Hayashi Y, Harashima H, Ohkohchi N. Protecting liver sinusoidal endothelial cells suppresses apoptosis in acute liver damage. Hepatol Res 2016; 46:697-706. [PMID: 26490536 DOI: 10.1111/hepr.12607] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/30/2015] [Accepted: 10/14/2015] [Indexed: 01/01/2023]
Abstract
AIM Apoptosis is associated with various types of hepatic disorders. We have developed a novel cell-transfer drug delivery system (DDS) using a multifunctional envelope-type nano device that targets liver sinusoidal endothelial cells (LSECs). The purpose of this study was to determine the efficacy of the novel DDS containing siRNA at suppressing apoptosis in LSECs. METHODS Bax siRNA was transfected into a sinusoidal endothelial cell line (M1) to suppress apoptosis induced by an anti-Fas antibody and staurosporine. C57BL/6J mice were divided into three groups: (i) a control group, only intravenous saline; (ii) a nonselective group, injections of siRNA sealed in the nonselective DDS; and (iii) an LSEC-transfer efficient group, injections of siRNA sealed in an LSEC-transfer efficient DDS. Hepatic cell apoptosis was induced by an anti-Fas antibody. RESULTS Bax siRNA had an anti-apoptotic effect on M1 cells. Serum alanine aminotransferase was reduced in the LSEC-transfer efficient group, as were cleaved caspase-3 and the number of terminal deoxynucleotidyl transferase dUTP nick end labeling positive hepatocytes. Silver impregnation staining indicated that the sinusoidal space was maintained in the LSEC-transfer efficient group but not in the other groups. Electron microscopy showed that the LSECs were slightly impaired, although the sinusoidal structure was maintained in the LSEC-transfer efficient group. CONCLUSION Hepatocyte apoptosis was reduced by the efficient suppression of LSEC apoptosis with a novel DDS. Protecting the sinusoidal structure by suppressing LSEC damage will be an effective treatment for acute liver failure.
Collapse
Affiliation(s)
- Tomohito Tanoi
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takafumi Tamura
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Naoki Sano
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Ken Nakayama
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kiyoshi Fukunaga
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yun-Wen Zheng
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Afsana Akhter
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yu Sakurai
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yasuhiro Hayashi
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Nobuhiro Ohkohchi
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
11
|
Wajant H. Principles of antibody-mediated TNF receptor activation. Cell Death Differ 2015; 22:1727-41. [PMID: 26292758 PMCID: PMC4648319 DOI: 10.1038/cdd.2015.109] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/26/2015] [Accepted: 07/01/2015] [Indexed: 12/17/2022] Open
Abstract
From the beginning of research on receptors of the tumor necrosis factor (TNF) receptor superfamily (TNFRSF), agonistic antibodies have been used to stimulate TNFRSF receptors in vitro and in vivo. Indeed, CD95, one of the first cloned TNFRSF receptors, was solely identified as the target of cell death-inducing antibodies. Early on, it became evident from in vitro studies that valency and Fcγ receptor (FcγR) binding of antibodies targeting TNFRSF receptors can be of crucial relevance for agonistic activity. TNFRSF receptor-specific antibodies of the IgM subclass and secondary cross-linked or aggregation prone dimeric antibodies typically display superior agonistic activity compared with dimeric antibodies. Likewise, anchoring of antibodies to cell surface-expressed FcγRs potentiate their ability to trigger TNFRSF receptor signaling. However, only recently has the relevance of oligomerization and FcγR binding for the in vivo activity of antibody-induced TNFRSF receptor activation been straightforwardly demonstrated in vivo. This review discusses the crucial role of oligomerization and/or FcγR binding for antibody-mediated TNFRSF receptor stimulation in light of current models of TNFRSF receptor activation and especially the overwhelming relevance of these issues for the rational development of therapeutic TNFRSF receptor-targeting antibodies.
Collapse
Affiliation(s)
- H Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
12
|
A novel hypothesis for an alkaline phosphatase 'rescue' mechanism in the hepatic acute phase immune response. Biochim Biophys Acta Mol Basis Dis 2013; 1832:2044-56. [PMID: 23899605 DOI: 10.1016/j.bbadis.2013.07.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 07/10/2013] [Accepted: 07/22/2013] [Indexed: 12/24/2022]
Abstract
The liver isoform of the enzyme alkaline phosphatase (AP) has been used classically as a serum biomarker for hepatic disease states such as hepatitis, steatosis, cirrhosis, drug-induced liver injury, and hepatocellular carcinoma. Recent studies have demonstrated a more general anti-inflammatory role for AP, as it is capable of dephosphorylating potentially deleterious molecules such as nucleotide phosphates, the pathogenic endotoxin lipopolysaccharide (LPS), and the contact clotting pathway activator polyphosphate (polyP), thereby reducing inflammation and coagulopathy systemically. Yet the mechanism underlying the observed increase in liver AP levels in circulation during inflammatory insults is largely unknown. This paper hypothesizes an immunological role for AP in the liver and the potential of this system for damping generalized inflammation along with a wide range of ancillary pathologies. Based on the provided framework, a mechanism is proposed in which AP undergoes transcytosis in hepatocytes from the canalicular membrane to the sinusoidal membrane during inflammation and the enzyme's expression is upregulated as a result. Through a tightly controlled, nucleotide-stimulated negative feedback process, AP is transported in this model as an immune complex with immunoglobulin G by the asialoglycoprotein receptor through the cell and secreted into the serum, likely using the receptor's State 1 pathway. The subsequent dephosphorylation of inflammatory stimuli by AP and uptake of the circulating immune complex by endothelial cells and macrophages may lead to decreased inflammation and coagulopathy while providing an early upstream signal for the induction of a number of anti-inflammatory gene products, including AP itself.
Collapse
|
13
|
Chan DV, Sharma R, Ju CYA, Roffler SR, Ju ST. A recombinant scFv-FasLext as a targeting cytotoxic agent against human Jurkat-Ras cancer. J Biomed Sci 2013; 20:16. [PMID: 23497165 PMCID: PMC3599744 DOI: 10.1186/1423-0127-20-16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 02/22/2013] [Indexed: 11/17/2022] Open
Abstract
Background Targeted therapy of human cancers is an attractive approach and has been investigated with limited success. We have developed novel cytotoxic agents for targeted therapy of human cancers based on the extracellular cytotoxicity domain of CD178 (FasL) and the specificity offered by single chain antibodies (scFv) against dominant human tumor Ag TAG-72 (cc49scFv) and TAL6 (L6scFv). Results The cc49scFv-FasLext is highly effective in in vitro killing of human TAG-72+ Jurkat-Ras tumor cells with a 30,000 fold greater cytotoxicity as compared to soluble FasL (sFasL). On the other hand, L6scFv-FasLext only increased cytotoxicity 500-fold as compared with sFasL against TAL6+ HeLa cells in in vitro assays. The high specificity and strong cytotoxicity of cc49scFv-FasLext made it feasible to cure IP-implanted Jurkat-Ras tumors in SCID mice. Conclusion Our study demonstrated that scFv-FasLext with a strong cytotoxicity against sensitive human tumor targets may be useful as effective chemotherapeutic agents.
Collapse
Affiliation(s)
- Derek V Chan
- Department of Pathology, Boston University School of Medicine, Boston University, Boston, MA 02118, USA
| | | | | | | | | |
Collapse
|
14
|
Yan XF, Liu P, Sun MY, Wang XL. Mechanisms underlying the therapeutic effect of Huangqi Decoction against dimethylnitrosamine-induced liver fibrosis in rats. Shijie Huaren Xiaohua Zazhi 2010; 18:2410-2415. [DOI: 10.11569/wcjd.v18.i23.2410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the mechanisms underlying the therapeutic effect of Huangqi Decoction against dimethylnitrosamine (DMN)-induced liver fibrosis in rats.
METHODS: Liver fibrosis was induced in rats by intraperitoneal injection of DMN for 4 wk. Rats were randomly divided into two groups: normal group and model group. Fibrotic rats in the model group were further randomly divided into two subgroups: model control subgroup and Huangqi Decoction subgroup. The Huangqi Decoction subgroup was intragastrically administered Huangqi Decoction for 2 wk, while the model control subgroup was administered equal volume of saline. At the end of 2, 4 and 6 wk, hepatic tissue samples were collected to detect the protein expression of Fas, caspase-8, caspase-3, matrix metallopeptidase-9 (MMP-9), tissue inhibitor of metalloproteinase 1 (TIMP-1) and TIMP-2 by Western blot, mRNA expression of α-SMA by real time-PCR, and MMP-2 and MMP-9 activity by gelatin enzymography.
RESULTS: Compared with the normal group, the expression levels of Fas, caspase-8, caspase-3, TIMP-1 and TIMP-2 proteins and α-SMA mRNA as well as MMP-2 and MMP-9 activity in liver tissue increased gradually in the model group and peaked at 4 wk. Compared with the model control subgroup, the expression levels of Fas, caspase-8, caspase-3, TIMP-1 and TIMP-2 proteins and α-SMA mRNA as well as MMP-2 activity at 6 wk were significantly reduced (1.05 ± 0.02 vs 1.17 ± 0.04, 1.41 ± 0.04 vs 1.98 ± 0.06, 0.86 ± 0.01 vs 1.19±0.04, 1.03 ± 0.03 vs 1.58 ± 0.06, 1.16 ± 0.04 vs 1.53 ± 0.01, 3.12 ± 0.47 vs 8.48 ± 0.45 and 2.15 ± 0.03 vs 2.33 ± 0.05, respectively; all P < 0.05 or 0.01), and MMP-9 protein expression and activity were significantly increased (1.21 ± 0.00 vs 1.12 ± 0.01 and 1.25 ± 0.07 vs 1.10 ± 0.04, respectively; both P < 0.05 or 0.01) in liver tissue in the Huangqi Decoction subgroup.
CONCLUSION: Huangqi Decoction exerts significant anti-fibrotic effects perhaps by inhibiting hepatic cell apoptosis and hepatic stellate cell (HSC) activation, modulating the MMPs/TIMPs system, and promoting extracellular matrix (ECM) degradation.
Collapse
|
15
|
Yoshida H, Watanabe K, Takahashi S, Ichikawa K. Protective effects of HFE7A, mouse anti-human/mouse Fas monoclonal antibody against acute and lethal hepatic injury induced by Jo2. Cytotechnology 2009; 62:313-23. [PMID: 20024619 DOI: 10.1007/s10616-009-9244-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Accepted: 12/02/2009] [Indexed: 01/06/2023] Open
Abstract
HFE7A is a mouse anti-human/mouse Fas monoclonal antibody which, protects mice from fulminant hepatitis induced by Jo2. Herein, we report on the mechanism of the protective effect of HFE7A against Jo2-induced acute and lethal hepatic injury. HFE7A reduced the serum aminotransferase level which was elevated after Jo2 injection. HFE7A also inhibited caspase activation and mitochondrial depolarization in hepatocytes derived from apoptosis induced by Jo2 injection. The protective effect of HFE7A against Jo2-induced apoptosis in mouse hepatocytes was reproducible in vitro. The cell death and caspase activation in isolated mouse hepatocytes were induced by incubating these cells with Jo2 in vitro, and HFE7A inhibited the cell death and caspase activation in mouse hepatocytes in a dose-dependent manner. The affinity of HFE7A to mouse Fas was lower than that of Jo2. The binding of Jo2 to neither recombinant mouse Fas nor mouse hepatocytes was inhibited by an excessive amount of HFE7A. Interestingly, HFE7A bound to hepatocytes isolated from Fas knockout mice. From these results, it is suggested that HFE7A may exert a protective effect against Jo2-induced hepatitis not by competitively inhibiting the binding of Jo2 to Fas on hepatocytes, and that a distinct molecule other than Fas may possibly be involved in the protective effect of HFE7A against Jo2-induced hepatic injury.
Collapse
Affiliation(s)
- Hiroko Yoshida
- Biological Research Laboratories IV, Daiichi Sankyo Co., Ltd., Tokyo, 134-8630, Japan,
| | | | | | | |
Collapse
|
16
|
Jiang L, Li JG, Lan L, Wang YM, Liu GD. Repopulation of human fetal hepatocytes in nude mouse model with chimeric human liver using mouse-specific anti-Fas antibody. Shijie Huaren Xiaohua Zazhi 2008; 16:1734-1740. [DOI: 10.11569/wcjd.v16.i16.1734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate repopulation of human fetal hepatocytes in an animal model of nude mice with chimeric human liver following induction of mouse hepatocyte apoptosis using a mouse-specific anti-Fas monoclonal antibody (Jo2 mAb) that does not engage xenogeneic fas.
METHODS: For experiment group, nude mice were transplanted with human fetal hepatocytes intrasplenically and treated with 0.2 mg/kg Jo2 mAb intraperitoneally once a week for 12 weeks consistently. Nude mice in the control group were transplanted with human fetal hepatocytes but not administrated with Jo2 mAb. Liver section from non-transplanted nude mice administered with Jo2 mAb were analyzed using hematoxylin and eosin staining and terminal uridine deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. Reverse transcription-polymerase chain reaction (RT-PCR) and S-P immunohistochemistry were used to detect human albumin mRNA, human albumin and specific proliferating cell nuclear antigen (PCNA) in chimeric liver tissues.
RESULTS: Liver sections from non-transplanted nude mice administered with Jo2 mAb showed hepatocyte death, massive apoptosis and hemorrhage. Nude mice in both experiment group and control group survived 24 weeks after transplantation. Human albumin and specific human PCNA were detected from the week 2 to week 20 after transplantation, but they could only be detected from the week 2 to week 12 in the controls. Human albumin mRNA (356 bp) was detected in mice livers from the week 4 to week 16 after transplantation, but they could only be detected from the week 4 to week 8 in the controls. The number of PCNA in experiment group is significantly higher than in the control group at 8, 12 wk (25.7% ± 8.5% vs 13.4% ± 7.8%, 29.4%± 5.0% vs 8.5% ± 2.3%, both P < 0.05).
CONCLUSION: Human fetal hepatocytes of xenogeneic graft can survive in nude mice. The repopulation of human fetal hepatocytes can be promoted and prolonged in nude mouse model with chimeric human liver using mouse-specific anti-Fas antibody intraperitoneally.
Collapse
|
17
|
Newkirk MM, Nowak U, Skamene E, Iera D, Desbarats J. Agonistic antibodies to Fas induce a breach in the endothelial lining of the liver and a breakdown in B cell tolerance. Clin Exp Immunol 2007; 147:346-51. [PMID: 17223977 PMCID: PMC1810459 DOI: 10.1111/j.1365-2249.2006.03279.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Liver disease can be associated with a breakdown in self-tolerance and the production of autoantibodies such as rheumatoid factors (RF), which bind to IgG. Here we investigated whether primary, non-infectious liver damage was sufficient to induce autoantibody production. We established a model of targeted liver damage induced by weekly sublethal injections of pro-apoptotic anti-Fas (CD95) antibodies. Liver damage, monitored by measurements of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, was minimal 1 week after anti-Fas injection. However, the sublethal Fas stimulation was sufficient to trigger significant haemorrhage in the liver, as assessed by Evans Blue dye leakage into the organ 5 h after anti-Fas antibody injection. We observed an induction of RF in response to the weekly injections of sublethal anti-Fas antibodies but not of isotype control antibodies, indicating a breakdown of self-tolerance induced by Fas engagement. RF induction was unlikely to be due to direct activation of B cells, as splenocytes stimulated with anti-Fas antibodies in vitro did not produce RF. These studies show that sublethal damage to the liver by Fas engagement leads to liver haemorrhage and is sufficient to trigger the breakdown of self-tolerance.
Collapse
Affiliation(s)
- M M Newkirk
- Research Institute of McGill University Health Centre, Department of Medicine, Montreal, Quebec, Canada.
| | | | | | | | | |
Collapse
|
18
|
Leifeld L, Fink K, Debska G, Fielenbach M, Schmitz V, Sauerbruch T, Spengler U. Anti-apoptotic function of gelsolin in fas antibody-induced liver failure in vivo. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:778-85. [PMID: 16507893 PMCID: PMC1606525 DOI: 10.2353/ajpath.2006.050323] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Apoptosis is a key mechanism underlying fulminant hepatic failure. The role of gelsolin in such apoptotic pathways is not well understood because both pro-apoptotic and anti-apoptotic effects have been reported in vitro, depending on the cell type and in vitro expression model used. Therefore, we studied an in vivo model of hepatic failure by analyzing expression of gelsolin; intrahepatic activation of caspase-3, -8, and -9; and the extent of apoptosis in gelsolin knockout (gsn(-/-)) versus wild-type mice (gsn(+/+)) after exposure to stimulatory Fas antibody Jo-2. Gelsolin was expressed exclusively in sinusoidal lining cells, including sinusoidal endothelial cells and Kupffer cells, of gsn(+/+) mice. Compared with wild-type mice, Jo2-exposed gsn(-/-) mice showed significantly higher numbers of apoptotic cells in the liver (22 +/- 9 versus 5 +/- 4% terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling-positive cells, P = 0.002); shorter survival (P = 0.037); and enhanced activation of caspase-3 (P = 0.009), -8 (P = 0.004), and -9 (P = 0.004). Furthermore, inhibition of caspase-3 with z-DEVD-fmk blocked Jo2-induced liver failure in all mice. Thus, our data on Jo2-induced hepatic failure suggest that gelsolin exerts an overall anti-apoptotic effect in vivo. Moreover, selective expression of gelsolin in sinusoidal endothelial cells indicates a pivotal role for interactions between sinusoidal endothelial cells and liver parenchymal cells in Fas ligand-mediated liver failure.
Collapse
Affiliation(s)
- Ludger Leifeld
- Departments of Internal Medicine I, University of Bonn, Bonn, Germany.
| | | | | | | | | | | | | |
Collapse
|
19
|
Spierings DC, de Vries EG, Vellenga E, van den Heuvel FA, Koornstra JJ, Wesseling J, Hollema H, de Jong S. Tissue distribution of the death ligand TRAIL and its receptors. J Histochem Cytochem 2004; 52:821-31. [PMID: 15150291 DOI: 10.1369/jhc.3a6112.2004] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Recombinant human (rh) TNF-related apoptosis-inducing ligand (TRAIL) harbors potential as an anticancer agent. RhTRAIL induces apoptosis via the TRAIL receptors TRAIL-R1 and TRAIL-R2 in tumors and is non-toxic to nonhuman primates. Because limited data are available about TRAIL receptor distribution, we performed an immunohistochemical (IHC) analysis of the expression of TRAIL-R1, TRAIL-R2, the anti-apoptotic TRAIL receptor TRAIL-R3, and TRAIL in normal human and chimpanzee tissues. In humans, hepatocytes stained positive for TRAIL and TRAIL receptors and bile duct epithelium for TRAIL, TRAIL-R1, and TRAIL-R3. In brains, neurons expressed TRAIL-R1, TRAIL-R2, TRAIL-R3 but no TRAIL. In kidneys, TRAIL-R3 was negative, tubuli contorti expressed TRAIL-R1, TRAIL-R2, and TRAIL, and cells in Henle's loop expressed only TRAIL-R2. Heart myocytes showed positivity for all proteins studied. In colon, TRAIL-R1, TRAIL-R2, and TRAIL were present. Germ and Leydig cells were positive for all proteins studied. Endothelium in liver, heart, kidney, and testis lacked TRAIL-R1 and TRAIL-R2. In alveolar septa and bronchial epithelium TRAIL-R2 was expressed, brain vascular endothelium expressed TRAIL-R2 and TRAIL-R3, and in heart vascular endothelium only TRAIL-R3 was present. Only a few differences were observed between human and chimpanzee liver, brain, and kidney. In contrast to human, chimpanzee bile duct epithelium lacked TRAIL, TRAIL-R1, and TRAIL-R3, lung and colon showed no TRAIL or its receptors, TRAIL-R3 was absent in germ and Leydig cells, and vascular endothelium showed only TRAIL-R2 expression in the brain. In conclusion, comparable expression of TRAIL and TRAIL receptors was observed in human and chimpanzee tissues. Lack of liver toxicity in chimpanzees after rhTRAIL administration despite TRAIL-R1 and TRAIL-R2 expression is reassuring for rhTRAIL application in humans.
Collapse
Affiliation(s)
- Diana C Spierings
- Department of Medical Oncology, University Hospital Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Antigen-specific lymphocytes are involved in synovial proliferation within inflamed joints. Activated lymphocytes and synoviocytes from patients with rheumatoid arthritis express receptors that can bind TNF-related apoptosis-inducing ligand (TRAIL). A new study demonstrates that DCs pulsed with collagen and transduced with an adenovirus-based vector able to express TRAIL limit the incidence of arthritis in a model of collagen-induced arthritis and joint inflammation. These results suggest that gene-modified cell therapy represents a therapeutic option for systemic rheumatic diseases.
Collapse
Affiliation(s)
- George C Tsokos
- Walter Reed Army Institute of Research, Building 503, Room 1A32, Robert Grand Road, Silver Spring, Maryland 20910, USA.
| | | |
Collapse
|
21
|
|