1
|
Zhao B, Li X, Li X, Quan D, Zhang F, Hu B. Ilomastat contributes to the survival of mouse after irradiation via promoting the recovery of hematopoietic system. PLoS One 2021; 16:e0238209. [PMID: 33513143 PMCID: PMC7845947 DOI: 10.1371/journal.pone.0238209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 12/19/2020] [Indexed: 11/21/2022] Open
Abstract
Ilomastat, a broad-spectrum inhibitor of matrix metalloproteinases (MMPs), has drawn attentions for its function in alleviating radiation damage. However, the detailed mechanisms of Ilomastat’s protection from animal model remain not fully clear. In this study, the C57BL/6 mice were pre-administrated with Ilomastat or vihicle for 2 h, and then total body of mice were exposed to 6 Gy of γ-rays. The protective effect of Ilomastat on the hematopoietic system in the irradiated mice were investigated. We found that pretreatment with Ilomastat significantly reduced the level of TGF-β1 and TNF-α, and elevated the number of bone marrow (BM) mononuclear cells in the irradiated mice. Ilomastat pretreatment also increased the fraction of BM hematopoietic progenitor cells (HPCs) and hematopoietic stem cells (HSCs) at day 30 after irradiation, and protected the spleen of mouse from irradiation. These results suggest that Ilomastat promotes the recovery of hematopoietic injury in the irradiated mice, and thus contributes to the survival of mouse after irradiation.
Collapse
Affiliation(s)
- Baoquan Zhao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
- * E-mail:
| | - Xiaoman Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Department of Radiation Medicine, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xingzhou Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
| | - Dongqin Quan
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
| | - Fang Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
| | - Burong Hu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| |
Collapse
|
2
|
Minond D. Novel Approaches and Challenges of Discovery of Exosite Modulators of a Disintegrin and Metalloprotease 10. Front Mol Biosci 2020; 7:75. [PMID: 32435655 PMCID: PMC7218085 DOI: 10.3389/fmolb.2020.00075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
A disintegrin and metaproteinase 10 is an important target for multiple therapeutic areas, however, despite drug discovery efforts by both industry and academia no compounds have reached the clinic so far. The lack of enzyme and substrate selectivity of developmental drugs is believed to be a main obstacle to the success. In this review, we will focus on novel approaches and associated challenges in discovery of ADAM10 selective modulators that can overcome shortcomings of previous generations of compounds and be translated into the clinic.
Collapse
Affiliation(s)
- Dmitriy Minond
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Fort Lauderdale, FL, United States.,Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| |
Collapse
|
3
|
Nicolaou A, Zhao Z, Northoff BH, Sass K, Herbst A, Kohlmaier A, Chalaris A, Wolfrum C, Weber C, Steffens S, Rose-John S, Teupser D, Holdt LM. Adam17 Deficiency Promotes Atherosclerosis by Enhanced TNFR2 Signaling in Mice. Arterioscler Thromb Vasc Biol 2016; 37:247-257. [PMID: 28062509 DOI: 10.1161/atvbaha.116.308682] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 11/28/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVE ADAM17 (a disintegrin and metalloproteinase 17) is a sheddase releasing different types of membrane-bound proteins, including adhesion molecules, cytokines, and their receptors as well as inflammatory mediators. Because these substrates modulate important mechanisms of atherosclerosis, we hypothesized that ADAM17 might be involved in the pathogenesis of this frequent disease. APPROACH AND RESULTS Because Adam17-knockout mice are not viable, we studied the effect of Adam17 deficiency on atherosclerosis in Adam17 hypomorphic mice (Adam17ex/ex), which have low residual Adam17 expression. To induce atherosclerosis, mice were crossed onto the low-density lipoprotein receptor (Ldlr)-deficient background. We found that Adam17ex/ex.Ldlr-/- mice developed ≈1.5-fold larger atherosclerotic lesions, which contained more macrophages and vascular smooth muscle cells than wild-type littermate controls (Adam17wt/wt.Ldlr-/-). Reduced Adam17-mediated shedding led to significantly increased protein levels of membrane-resident TNFα (tumor necrosis factor) and TNFR2 (tumor necrosis factor receptor 2), resulting in a constitutive activation of TNFR2 signaling. At the same time, Adam17 deficiency promoted proatherosclerotic cellular functions, such as increased proliferation and reduced apoptosis in cultured macrophages and vascular smooth muscle cells and increased adhesion of macrophages to vascular endothelial cells. Because siRNA (small interfering RNA)-mediated knockdown of Tnfr2 rescued from aberrant proliferation and from misregulation of apoptosis in Adam17-depleted cells, our data indicate that TNFR2 is an important effector of ADAM17 in our mouse model. CONCLUSIONS Our results provide evidence for an atheroprotective role of ADAM17, which might be mediated by cleaving membrane-bound TNFα and TNFR2, thereby preventing overactivation of endogenous TNFR2 signaling in cells of the vasculature.
Collapse
Affiliation(s)
- Alexandros Nicolaou
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Zhen Zhao
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Bernd H Northoff
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Kristina Sass
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Andreas Herbst
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Alexander Kohlmaier
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Athena Chalaris
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Christian Wolfrum
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Christian Weber
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Sabine Steffens
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Stefan Rose-John
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Daniel Teupser
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Lesca M Holdt
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.).
| |
Collapse
|
4
|
Bahador M, Cross AS. Review: From therapy to experimental model: a hundred years of endotoxin administration to human subjects. ACTA ACUST UNITED AC 2016; 13:251-79. [DOI: 10.1177/0968051907085986] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This article is a review of studies in which endotoxin has been administered to human subjects for experimental purposes. Data are presented in tabular form so the reader can better appreciate the objectives of individual studies. Although the original intention was to focus on the adverse events associated with these studies, unexpected serious adverse events rarely have been reported.
Collapse
Affiliation(s)
- Marjan Bahador
- Center for Vaccine Development and Division of Infectious Diseases, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA,
| | - Alan S. Cross
- Center for Vaccine Development and Division of Infectious Diseases, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Hoffmann E, Wald J, Lavu S, Roberts J, Beaumont C, Haddad J, Elliott P, Westphal C, Jacobson E. Pharmacokinetics and tolerability of SRT2104, a first-in-class small molecule activator of SIRT1, after single and repeated oral administration in man. Br J Clin Pharmacol 2013; 75:186-96. [PMID: 22616762 DOI: 10.1111/j.1365-2125.2012.04340.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 05/12/2012] [Indexed: 12/15/2022] Open
Abstract
AIM SRT2104 is a novel, first-in-class, highly selective small molecule activator of the NAD + dependent deacetylase SIRT1. SRT2104 was dosed to healthy male and female volunteers in a series of phase 1 clinical studies that were designed to elucidate tolerability and pharmacokinetics associated with oral dosing to aid in dose selection for subsequent clinical trials. METHODS In the first-in-human study, there was both a single dose phase and 7 day repeat dose phase. Doses used ranged from 0.03 to 3.0 g. A radioactive microtracer study was subsequently conducted to determine systemic clearance, bioavailability and preliminary metabolism, and a crossover study was conducted to determine the effect of gender, formulation and feeding state on SRT2104 pharmacokinetics. RESULTS SRT2104 was well tolerated in all of these studies, with no serious adverse reactions observed. SRT2104 displayed a dose-dependent, but sub-proportional increase in exposure following single dose and repeated dose administration. Accumulation of three-fold or less occurs after 7 days of repeat dosing. The mean bioavailability was circa 14% and the mean clearance was circa 400 ml min(-1). Although there were no substantial effects on exposure resulting from gender or formulation differences, a notable food effect was observed, manifested as up to four-fold increase in exposure parameters. CONCLUSIONS In the absence of an optimized formulation of SRT2104, the food effect can be used to maximize exposure in future clinical studies. Combined with the good tolerability of all doses demonstrated in these studies, the favourable selectivity profile of SRT2104 allows for the use of this SIRT1 modulator for target validation in the clinic.
Collapse
|
6
|
Sharma M, Mohapatra J, Acharya A, Deshpande SS, Chatterjee A, Jain MR. Blockade of tumor necrosis factor-α converting enzyme (TACE) enhances IL-1β and IFN-γ via caspase-1 activation: a probable cause for loss of efficacy of TACE inhibitors in humans? Eur J Pharmacol 2012; 701:106-13. [PMID: 23266381 DOI: 10.1016/j.ejphar.2012.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 12/01/2012] [Accepted: 12/07/2012] [Indexed: 10/27/2022]
Abstract
TNF-α converting enzyme (TACE) is a member of the ADAM (a disintegrin and metalloproteinase) family and is known as ADAM17, which processes precursor TNF-α in order to release soluble TNF-α (sTNF-α). Inhibition of TACE has been effective as a strategy to inhibit arthritis in animal models; however, it has not been translated in the clinic due to lack of efficacy or toxicity. We hypothesized that inhibition of TACE may activate a different pro-inflammatory pathway in human. To investigate this, we studied the effect of TACE inhibitor DPC-333 on cytokine levels in concanavalin A (Con A) activated human peripheral blood mononuclear cells (hPBMC). We have also studied the effects of DPC-333 on Con A induced cytokine levels in mice in vivo or in vitro in whole blood assay. DPC-333 treatment significantly up-regulated IL-1β and IFN-γ in Con A activated hPBMC. In contrast, pre-treatment with DPC-333 effectively suppressed IL-1β and IFN-γ in mice in vivo or in vitro. Interestingly, DPC-333 was found to up-regulate mRNA expression of caspase-1 in hPBMC in a dose dependent fashion and selective caspase-1 inhibitor completely restored DPC-333 induced IL-1β and IFN-γ. Furthermore, selective IL-1β receptor antagonist (anakinra) prevented DPC-333 induced IFN-γ. In conclusion, our data demonstrates that blockade of TACE enhances IL-1β in a caspase-1 dependent manner in vitro in hPBMC and the elevation of IFN-γ is secondarily mediated via IL-1β. This novel finding might explain the possible cause behind the loss of efficacy of TACE inhibitors in human.
Collapse
Affiliation(s)
- Manoranjan Sharma
- Department of Pharmacology, Zydus Research Centre, Moraiya, Ahmedabad, Gujarat, India
| | | | | | | | | | | |
Collapse
|
7
|
Kelly CJ, Colgan SP, Frank DN. Of microbes and meals: the health consequences of dietary endotoxemia. Nutr Clin Pract 2012; 27:215-25. [PMID: 22378797 DOI: 10.1177/0884533611434934] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The human intestinal tract comprises a rich and complex microbial ecosystem. This intestinal microbota provides a large reservoir of potentially toxic molecules, including bacterial endotoxin (ie, lipopolysaccharide [LPS]). This potent inflammatory molecule is detectable in the circulation of healthy individuals, and levels transiently increase following ingestion of energy-rich meals. Chronic exposure to circulating endotoxin has been associated with obesity, diabetes, and cardiovascular disease. Western-style meals augment LPS translocation and by this mechanism may contribute to the pathogenesis of these diseases. By contrast, the gut and other organs have evolved mechanisms to detoxify endotoxin and neutralize the potentially inflammatory qualities of circulating endotoxin. Of specific interest to clinicians is evidence that acute postprandial elevation of circulating endotoxin is dependent on meal composition. In this review, the authors present an overview of the biochemical and cellular mechanisms that lead to endotoxemia, with emphasis on the interplay between microbial and nutrition determinants of this condition. The link between endotoxemia, diet, and changes in the intestinal microbiota raise the possibility that dietary interventions can, at least in part, ameliorate the detrimental outcomes of endotoxemia.
Collapse
Affiliation(s)
- Caleb J Kelly
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | | | | |
Collapse
|
8
|
Shu C, Zhou H, Afsharvand M, Duan L, Zhang H, Noveck R, Raible D. Pharmacokinetic-pharmacodynamic modeling of apratastat: a population-based approach. J Clin Pharmacol 2010; 51:472-81. [PMID: 21059888 DOI: 10.1177/0091270010372389] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Apratastat is an orally active, potent, and reversible dual inhibitor of tumor necrosis factor-α converting enzyme (TACE) and matrix metalloproteinases (MMPs). This study characterizes the pharmacodynamic (PD) effect of apratastat following oral administration on tumor necrosis factor-alpha (TNF-α) release. Data were obtained from 3 clinical studies carried out in healthy subjects. Apratastat was administered orally in these studies as single doses or multiple doses (twice daily). The inhibition of TNF-α release by apratastat was investigated in studies of in vitro, ex vivo, and in vivo. Inhibitory E(max) models were used to characterize the inhibition of TNF-α release in both in vitro and ex vivo studies. Apratastat inhibited TNF-α release with a population mean IC(50) of 144 ng/mL in vitro and of 81.7 ng/mL ex vivo, respectively. The relationship between TNF-α and apratastat plasma concentration in the endotoxin-challenged study in healthy subjects was well characterized by a mechanism-based PD population model with IC(50) of 126 ng/mL. Apratastat can potently inhibit the release of TNF-α in vitro, ex vivo, and in vivo. Even though the dosage provided adequate exposure to inhibit TNF-α release, apratastat was not efficacious in rheumatoid arthritis (RA). This inconsistency between TNF-α inhibition and the clinical response requires further investigation.
Collapse
Affiliation(s)
- Cathye Shu
- Pfizer, 500 Arcola Road, Collegeville, PA 19426, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
van Goor H, Melenhorst WBWH, Turner AJ, Holgate ST. Adamalysins in biology and disease. J Pathol 2009; 219:277-86. [DOI: 10.1002/path.2594] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
10
|
Moss ML, Sklair-Tavron L, Nudelman R. Drug insight: tumor necrosis factor-converting enzyme as a pharmaceutical target for rheumatoid arthritis. ACTA ACUST UNITED AC 2008; 4:300-9. [PMID: 18414459 DOI: 10.1038/ncprheum0797] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Accepted: 02/19/2008] [Indexed: 12/13/2022]
Abstract
The success of agents that inhibit tumor necrosis factor (TNF), such as infliximab, adalimumab and etanercept, has led to a desire for orally available small molecules that have a better safety profile and are less costly to produce than current agents. One target for anti-TNF therapy that is currently under investigation is TNF-converting enzyme, which promotes the release of soluble TNF from its membrane-bound precursor. Inhibitors of this enzyme with drug-like properties have been made and tested in the clinic. These inhibitors include TMI-005 and BMS-561392, both of which have entered into phase II clinical trials. This article summarizes preclinical and clinical findings regarding the use of inhibitors of TNF-converting enzyme for the treatment of rheumatoid arthritis.
Collapse
|
11
|
Badellino KO, Wolfe ML, Reilly MP, Rader DJ. Endothelial Lipase Is Increased In Vivo by Inflammation in Humans. Circulation 2008; 117:678-85. [DOI: 10.1161/circulationaha.107.707349] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Karen O. Badellino
- From the School of Nursing (K.O.B.), Institute of Translational Medicine and Therapeutics (K.O.B., M.L.W., M.P.R., D.J.R.), School of Medicine (M.P.R., D.J.R.), and Cardiovascular Institute (M.P.R.), University of Pennsylvania, Philadelphia
| | - Megan L. Wolfe
- From the School of Nursing (K.O.B.), Institute of Translational Medicine and Therapeutics (K.O.B., M.L.W., M.P.R., D.J.R.), School of Medicine (M.P.R., D.J.R.), and Cardiovascular Institute (M.P.R.), University of Pennsylvania, Philadelphia
| | - Muredach P. Reilly
- From the School of Nursing (K.O.B.), Institute of Translational Medicine and Therapeutics (K.O.B., M.L.W., M.P.R., D.J.R.), School of Medicine (M.P.R., D.J.R.), and Cardiovascular Institute (M.P.R.), University of Pennsylvania, Philadelphia
| | - Daniel J. Rader
- From the School of Nursing (K.O.B.), Institute of Translational Medicine and Therapeutics (K.O.B., M.L.W., M.P.R., D.J.R.), School of Medicine (M.P.R., D.J.R.), and Cardiovascular Institute (M.P.R.), University of Pennsylvania, Philadelphia
| |
Collapse
|
12
|
Qian M, Bai SA, Brogdon B, Wu JT, Liu RQ, Covington MB, Vaddi K, Newton RC, Fossler MJ, Garner CE, Deng Y, Maduskuie T, Trzaskos J, Duan JJW, Decicco CP, Christ DD. Pharmacokinetics and pharmacodynamics of DPC 333 ((2R)-2-((3R)-3-amino-3{4-[2-methyl-4-quinolinyl) methoxy] phenyl}-2-oxopyrrolidinyl)-N-hydroxy-4-methylpentanamide)), a potent and selective inhibitor of tumor necrosis factor alpha-converting enzyme in rodents, dogs, chimpanzees, and humans. Drug Metab Dispos 2007; 35:1916-25. [PMID: 17656469 DOI: 10.1124/dmd.107.015933] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
DPC 333 ((2R)-2-((3R)-3-amino-3{4-[2-methyl-4-quinolinyl) methoxy] phenyl}-2-oxopyrrolidinyl)-N-hydroxy-4-methylpentanamide)) is a potent and selective inhibitor of tumor necrosis factor (TNF)-alpha-converting enzyme (TACE). It significantly inhibits lipopolysaccharide-induced soluble TNF-alpha production in blood from rodents, chimpanzee, and human, with IC(50) values ranging from 17 to 100 nM. In rodent models of endotoxemia, DPC 333 inhibited the production of TNF-alpha in a dose-dependent manner, with an oral ED(50) ranging from 1.1 to 6.1 mg/kg. Oral dosing of DPC 333 at 5.5 mg/kg daily for 2 weeks in a rat collagen antibody-induced arthritis model suppressed the maximal response by approximately 50%. DPC 333 was distributed widely to tissues including the synovium, the site of action for antiarthritic drugs. Pharmacokinetic and pharmacodynamic studies in chimpanzee revealed a systemic clearance of 0.4 l/h/kg, a V(ss) of 0.6 l/kg, an oral bioavailability of 17%, and an ex vivo IC(50) for the suppression of TNF-alpha production of 55 nM (n = 1). In a phase I clinical trial with male volunteers after single escalating doses of oral DPC 333, the terminal half-life was between 3 and 6 h and the ex vivo IC(50) for suppressing TNF-alpha production was 113 nM. Measurement of the suppression of TNF-alpha production ex vivo may serve as a good biomarker in evaluating the therapeutic efficacy of TACE inhibitors. Overall, the pharmacological profiles of DPC 333 support the notion that suppression of TNF-alpha with TACE inhibitors like DPC 333 may provide a novel approach in the treatment of various inflammatory diseases including rheumatoid arthritis, via control of excessive TNF-alpha production.
Collapse
Affiliation(s)
- Mingxin Qian
- Metabolism and Pharmacokinetics, Bristol-Myers Squibb Company, Princeton, New Jersey, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Alwayn IPJ, Andersson C, Lee S, Arsenault DA, Bistrian BR, Gura KM, Nose V, Zauscher B, Moses M, Puder M. Inhibition of matrix metalloproteinases increases PPAR-alpha and IL-6 and prevents dietary-induced hepatic steatosis and injury in a murine model. Am J Physiol Gastrointest Liver Physiol 2006; 291:G1011-9. [PMID: 16844679 DOI: 10.1152/ajpgi.00047.2006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Steatosis is a prominent feature of nonalcoholic fatty liver disease and a potential promoter of inflammation. Injury leading to cirrhosis is partly mediated by dysregulation of matrix protein turnover. Matrix metalloproteinase (MMP) inhibitors protect mice from lethal TNF-alpha induced liver injury. We hypothesized that Marimastat, a broad-spectrum MMP and TNF-alpha converting enzyme (TACE) inhibitor, might modulate this injury through interruption of inflammatory pathways. Triglyceride and phospholipid levels (liver, serum) and fatty acid profiles were used to assess essential fatty acid status and de novo lipogenesis as mechanisms for hepatic steatosis. Mice receiving a fat-free, high-carbohydrate diet (HCD) for 19 days developed severe fatty liver infiltration, demonstrated by histology, magnetic resonance spectroscopy, and elevated liver function tests. Animals receiving HCD plus Marimastat (HCD+MAR) were comparable to control animals. Increased tissue levels of peroxisome proliferator activated receptor-alpha (PPAR-alpha), higher levels of serum IL-6, and decreased levels of serum TNF-alpha receptor II were also seen in the HCD+MAR group compared with HCD-only. In addition, there was increased phosphorylation, and likely activation, of PPAR-alpha in the HCD+MAR group. PPAR-alpha is a transcription factor involved in beta-oxidation of fatty acids, and IL-6 is a hepatoprotective cytokine. Liver triglyceride levels were higher and serum triglyceride and phospholipid levels lower with HCD-only but improved with Marimastat treatment. HCD-only and HCD+MAR groups were essential fatty acid deficient and had elevated rates of de novo lipogenesis. We therefore conclude that Marimastat reduces liver triglyceride accumulation by increasing fat oxidation and/or liver clearance of triglycerides. This may be related to increased expression and activation of PPAR-alpha or IL-6, respectively.
Collapse
Affiliation(s)
- Ian P J Alwayn
- Department of Surgery and the Vascular Biology Program, Children's Hospital Boston, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Satoh M, Iwasaka J, Nakamura M, Akatsu T, Shimoda Y, Hiramori K. Increased expression of tumor necrosis factor-alpha converting enzyme and tumor necrosis factor-alpha in peripheral blood mononuclear cells in patients with advanced congestive heart failure. Eur J Heart Fail 2006; 6:869-75. [PMID: 15556048 DOI: 10.1016/j.ejheart.2004.02.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2003] [Revised: 12/19/2003] [Accepted: 02/23/2004] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Tumor necrosis factor-alpha converting enzyme (TACE) has recently been identified as a metalloproteinase-disintegrin, which converts pro-tumor necrosis factor-alpha (TNF-alpha) to the mature form, and is an important mediator in the pathogenesis of CHF. AIMS In order to establish the importance of TACE in the regulation of TNF-alpha synthesis in peripheral blood mononuclear cells (PBMC), we analyzed mRNAs and protein-positive cells of both TACE and TNF-alpha in PBMC obtained from patients with congestive heart failure (CHF). METHODS AND RESULTS PBMC were obtained from 46 patients with CHF and 22 controls. PBMC were activated by phorbol 12-myristate 13-acetate and ionomycin and assessed for TACE and TNF-alpha mRNAs by real-time RT-PCR, intracellular TACE and TNF-alpha levels by flow cytometry, and TNF-alpha secretion by supernatant ELISA. Levels of TACE and TNF-alpha mRNAs, intracellular TACE and TNF-alpha, and supernatant TNF-alpha were higher in CHF than in controls (P<0.001). There was a positive correlation between TACE and TNF-alpha levels in CHF patients (mRNA: r=0.60, P<0.001, intracellular protein levels: r=0.76, P<0.001). When the CHF group was divided into two subgroups by NYHA functional class (I and II vs. III and IV), levels of TACE and TNF-alpha were significantly higher in severe CHF patients (NYHA III or IV) than in mild CHF patients (NYHA I or II) (mRNA: P<0.001; intracellular protein levels: P<0.001). CONCLUSION These results demonstrate that in patients with CHF, and especially those with severe CHF, TACE expression in PBMC increases with TNF-alpha expression. These observations suggest that TACE in PBMC is an important regulator of TNF-alpha maturation, meaning that TACE may be a potential target for the inhibition of cellular TNF-alpha production in CHF.
Collapse
Affiliation(s)
- Mamoru Satoh
- Second Department of Internal Medicine, Iwate Medical University School of Medicine, Uchimaru 19-1, Morioka 020-8505, Iwate, Japan.
| | | | | | | | | | | |
Collapse
|
15
|
Edwards CK, Bendele AM, Reznikov LI, Fantuzzi G, Chlipala ES, Li L, Moldawer LL, Mountz JD, Li YYY, Dinarello CA. Soluble human p55 and p75 tumor necrosis factor receptors reverse spontaneous arthritis in transgenic mice expressing transmembrane tumor necrosis factor α. ACTA ACUST UNITED AC 2006; 54:2872-85. [PMID: 16947419 DOI: 10.1002/art.22077] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE The roles of the transmembrane and secreted forms of tumor necrosis factor alpha (TNFalpha) in rheumatoid arthritis (RA) remain unclear. Agents used to inhibit TNFalpha have shown varying efficacy in RA patients, suggesting that anti-TNFalpha agents possess dissimilar mechanisms of action, including the ability to neutralize transmembrane (tmTNFalpha) and secreted TNFalpha. In this study, TNFalpha-knockout (TNFalpha-KO) mice that were genetically altered to express elevated levels of tmTNFalpha were constructed to further understand the roles of the 17-kd secreted, trimeric, and 26-kd transmembrane forms of TNFalpha. METHODS A speed-congenic mating scheme was used to generate 3 unique strains of mice: 1) transgenic tmTgA86 mice overexpressing 26-kd tmTNFalpha and also secreting 17-kd trimeric TNFalpha (tmTNFalpha-transgenic), 2) TNFalpha-/- mice (TNFalpha-KO), and 3) transgenic mice overexpressing tmTNFalpha backcrossed to TNFalpha-KO mice (tmTNFalpha-transgenic/TNFalpha-KO). Mice were treated with phosphate buffered saline (as vehicle control), dexamethasone (as positive control), or modified recombinant human soluble TNF receptor (sTNFR) p55 or p75, and were assessed clinically and histopathologically for signs of inflammation and development of arthritis. RESULTS The tmTNFalpha-transgenic/TNFalpha-KO mice were born with crinkled tails and spinal deformities similar to those in ankylosing spondylitis. By 2-4 weeks, these mice developed symmetric inflammatory arthritis, characterized by tissue swelling, pannus formation, and bone deformities. The tmTNFalpha-transgenic mice also developed spontaneous-onset arthritis, but at a slower rate (100% incidence by 10-12 weeks). Clinical and histologic progression of arthritis in the tmTNFalpha-transgenic/TNFalpha-KO mice was reduced by treatment with dexamethasone or with the p55 or p75 sTNFR (69% and 63% reduction in total histologic score, respectively). CONCLUSION These data show that arthritis is sufficiently initiated and maintained in tmTNFalpha-transgenic/TNFalpha-KO mice, and that it can be neutralized by recombinant human p55 or p75 sTNFR, resulting in amelioration of the biologic and subsequent histologic destructive effects of tmTNFalpha.
Collapse
|
16
|
Marsik C, Jilma B, Joukhadar C, Mannhalter C, Wagner O, Endler G. The Toll-like receptor 4 Asp299Gly and Thr399Ile polymorphisms influence the late inflammatory response in human endotoxemia. Clin Chem 2005; 51:2178-80. [PMID: 16244296 DOI: 10.1373/clinchem.2005.051649] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Claudia Marsik
- Department of Clinical Pharmacology, Medical University of Vienna, Austria
| | | | | | | | | | | |
Collapse
|
17
|
Shiratsuch H, Basson MD. Differential regulation of monocyte/macrophage cytokine production by pressure. Am J Surg 2005; 190:757-62. [PMID: 16226954 DOI: 10.1016/j.amjsurg.2005.07.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2005] [Revised: 07/16/2005] [Accepted: 07/16/2005] [Indexed: 10/25/2022]
Abstract
BACKGROUND Cytokine production by macrophages is essential for the inflammatory response. Normal human interstitial tissue pressure is 20 to 30 mm Hg, but generally decreases in acute inflammation. METHODS We compared the effect of 20 mm Hg increased pressure (approximating normal interstitial tissue pressure) with that of ambient pressure (resembling pressure in inflamed tissues) on tumor necrosis factor (TNF)-alpha and interleukin (IL)-1beta production by undifferentiated (monocytic) and PMA (phorbol 12-, myristate 13-acetate)-differentiated (macrophage-like) THP-1 cells with or without lipopolysaccharide (LPS) (10 ng/mL). RESULTS Pressure stimulated spontaneous macrophage TNF-alpha secretion (30.5 +/- 6.3 vs. 49.1 +/- 2.8 pg/mL, P <.02), but not monocyte TNF-alpha secretion. Pressure did not stimulate IL-1beta release. As expected, LPS increased basal cytokine release. After LPS stimulation, pressure still tended to stimulate macrophage TNF-alpha, but inhibited monocyte TNF-alpha secretion (P <.05). In contrast, pressure inhibited IL-1beta release by both LPS-treated monocytes (986 +/- 134 vs. 595 +/- 226 pg/mL, P <.02) and macrophages (3,112 +/- 229 vs. 979 +/- 61 pg/mL, P <.01). CONCLUSIONS Extracellular pressure may regulate TNF-alpha and IL-1beta secretion differentially by monocytes and macrophages.
Collapse
|
18
|
Guler R, Olleros ML, Vesin D, Parapanov R, Garcia I. Differential effects of total and partial neutralization of tumor necrosis factor on cell-mediated immunity to Mycobacterium bovis BCG infection. Infect Immun 2005; 73:3668-76. [PMID: 15908396 PMCID: PMC1111814 DOI: 10.1128/iai.73.6.3668-3676.2005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The effects of total and partial inhibition of tumor necrosis factor (TNF) on sensitivity to Mycobacterium bovis BCG infection were investigated by using transgenic mice in which hepatocytes produced different amounts of human soluble TNF receptor 1 (sTNFR1) fused to the Fc fragment of human immunoglobulin G3 that could be detected in the serum. Transgenic mice expressing high serum levels of sTNFR1, neutralizing all circulating TNF, failed to develop differentiated granulomas and bactericidal mechanisms, and they succumbed to BCG infection. sTNFR1 transgenic mice did not activate BCG-induced Th1-type cytokines early in infection, but uncontrolled cytokine release was found late in infection. In this work we also evaluated the effect of partial inhibition of TNF on resistance to BCG infection. Transgenic mice expressing low levels of sTNFR1 were protected against BCG infection, and they developed increased bactericidal mechanisms, such as enhanced inducible nitric oxide synthase activity, increased macrophage activation, and showed higher numbers of liver granulomas early in infection compared to their negative littermates. Our data suggest that while total inhibition of TNF prevented BCG-induced cell-mediated immune responses, partial inhibition of TNF could contribute to macrophage activation, induction of bactericidal mechanisms, and granuloma formation in the early phase of BCG infection.
Collapse
Affiliation(s)
- Reto Guler
- Department of Pathology and Immunology, C.M.U., 1 rue Michel-Servet, CH 1211 Geneva 4, Switzerland
| | | | | | | | | |
Collapse
|
19
|
Zhang Y, Hegen M, Xu J, Keith JC, Jin G, Du X, Cummons T, Sheppard BJ, Sun L, Zhu Y, Rao VR, Wang Q, Xu W, Cowling R, Nickerson-Nutter CL, Gibbons J, Skotnicki J, Lin LL, Levin J. Characterization of (2R, 3S)-2-([[4-(2-butynyloxy)phenyl]sulfonyl]amino)-N,3-dihydroxybutanamide, a potent and selective inhibitor of TNF-alpha converting enzyme. Int Immunopharmacol 2005; 4:1845-57. [PMID: 15531300 DOI: 10.1016/j.intimp.2004.08.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2004] [Revised: 08/13/2004] [Accepted: 08/13/2004] [Indexed: 11/23/2022]
Abstract
TNF-alpha converting enzyme (TACE) is a validated therapeutic target for the development of oral tumor necrosis factor-alpha (TNF-alpha) inhibitors. Here we report the pre-clinical results and characterization of a selective and potent TACE inhibitor, (2R, 3S)-2-([[4-(2-butynyloxy)phenyl]sulfonyl]amino)-N,3-dihydroxybutanamide (TMI-2), in various in vitro and in vivo assays. TMI-2 is a potent TACE inhibitor in an enzymatic FRET assay (IC50=2 nM). It is more than 250-fold selective over MMP-1, -7, -9, -14, and ADAM-10 in vitro. In cell-based assays and human whole blood, TMI-2 inhibits lipopolysaccharide (LPS)-induced TNF secretion with IC50s<1 uM. Importantly, TMI-2 inhibits the spontaneous release of TNF-alpha in human synovium tissue explants of rheumatoid arthritis patients with an IC50 of 0.8 microM. In vivo, TMI-2 potently inhibits LPS-induced TNF-alpha production in mice (ED50=3 mg/kg). In the adjuvant-induced arthritis (AIA) model in rats, treatment with TMI-2 at 30 mg/kg and 100 mg/kg p.o. b.i.d. was highly effective in reducing joint arthritis scores. In a semi-therapeutic collagen-induced arthritis (CIA) model in mice, TMI-2 is highly effective in reducing disease severity scores after oral treatment at 100 mg/kg twice per day. In summary, TMI-2 is a potent and selective TACE inhibitor that inhibits TNF-alpha production and reduces the arthritis scores in pre-clinical models. TMI-2 represents a novel class of TACE inhibitors that may be effective and beneficial in the treatment of rheumatoid arthritis as well as other TNF-mediated inflammatory autoimmune diseases.
Collapse
Affiliation(s)
- Yuhua Zhang
- Department of Inflammation, Wyeth Research, 200 Cambridge Park Drive, Cambridge, MA 02140, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Shimoda Y, Satoh M, Nakamura M, Akatsu T, Hiramori K. Activated tumour necrosis factor-α shedding process is associated with in-hospital complication in patients with acute myocardial infarction. Clin Sci (Lond) 2005; 108:339-47. [PMID: 15603556 DOI: 10.1042/cs20040229] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
TACE [TNF-α (tumour necrosis factor-α)-converting enzyme] plays an essential role in the shedding of TNF-α, which could affect the outcome of AMI (acute myocardial infarction). To investigate the clinical significance of the TACE–TNF-α system in AMI, we examined TACE-mediated TNF-α synthesis in PBMCs (peripheral blood mononuclear cells), which are a possible source of TNF-α in AMI. Forty-one patients with AMI and 15 healthy subjects (HS) were enrolled in the present study. PBMCs were isolated from peripheral blood on day 1 and 14 after the onset of AMI. TACE and TNF-α mRNA levels and intracellular median fluorescence intensity were measured by real-time RT (reverse transcriptase)–PCR and flow cytometry respectively. TACE-mediated TNF-α production was evaluated in cultured PBMCs with PMA, which is known to activate TACE. Spontaneous TACE and TNF-α levels were higher in AMI patients than in HS (P<0.001). TACE and TNF-α levels in PMA-stimulated PMBCs were markedly increased in AMI patients compared with HS (P<0.001). There was a positive correlation between TACE and TNF-α levels in AMI. Although spontaneous and stimulated levels of TACE and TNF-α decreased 14 days after the onset of AMI, levels in AMI patients were higher than in HS. In AMI patients with in-hospital complications (n=15; pump failure in ten, recurrent myocardial infarction in one, malignant ventricular arrhythmia in three and cardiac death in one), spontaneous and stimulated levels of TACE and TNF-α were higher than in patients without complications (P<0.01). These levels were higher in AMI patients with in-hospital complications 14 days after onset. These results demonstrate that TACE-mediated TNF-α maturation in PBMCs may play an important role in poor outcomes from AMI, suggesting that TACE may be a potential target for the inhibition of cellular TNF-α production in AMI.
Collapse
Affiliation(s)
- Yudai Shimoda
- Second Department of Internal Medicine, Iwate Medical University School of Medicine, Uchimaru 19-1, Morioka 020-8505, Iwate, Japan
| | | | | | | | | |
Collapse
|
21
|
Abstract
Inflammatory bowel disease (IBD) is a chronic immunoinflammatory response to an stimulus that activates a chain of cellular mediators causing intestinal damage. One of the most well recognized proinflammatory mediators involved in the pathogenesis of IBD is tumor necrosis factor alpha (TNFalpha). The treatment of IBD has advanced in parallel to the improvement of the knowledge of its physiopathology, leading to the development of biological therapies. An example of this kind of treatment is the use of substances that antagonize TNFalpha, such as monoclonal antibodies infliximab, adalimumab, natalizumab, etanercept or onercept, with infliximab being the unique approved for use in IBD. Several studies have demonstrated that inhibition of TNFalpha is useful in the treatment of Crohn's disease (CD). In CD, infliximab induces the remission of relapses which are refractory to the conventional treatment, prevents more relapses and induces a closure of enterocutaneous and perianal fistula that do not respond to first line treatment. However, infliximab is not useful in ulcerative colitis. Infliximab treatment has some drawbacks, such as the development of anti-infliximab antibodies, which cause a loss of efficacy of the treatment and hypersensitivity reactions. Other reported adverse effects of infliximab are the development of autoimmunity, such as that related with antinuclear or anti-DNA antibodies, or the reactivation of infections such as tuberculosis. In fact, a screening for tuberculosis is necessary before administration of infliximab. To reduce the adverse effects due to infliximab immunogenicity, several trials with humanized or completely human agents, such as adalimumab or onercept, are under way. Until the precise stimulus that triggers IBD is identified, biological therapies have a great future and the selective antagonism of TNFalpha is already a reality.
Collapse
|
22
|
Corazza N, Brunner T, Buri C, Rihs S, Imboden MA, Seibold I, Mueller C. Transmembrane tumor necrosis factor is a potent inducer of colitis even in the absence of its secreted form. Gastroenterology 2004; 127:816-25. [PMID: 15362037 DOI: 10.1053/j.gastro.2004.06.036] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Tumor necrosis factor (TNF) is cleaved proteolytically from a 26-kilodalton transmembrane precursor protein into secreted 17-kilodalton monomers. Transmembrane (tm) and secreted trimeric TNF are biologically active and may mediate distinct activities. We assessed the consequences of a complete inhibition of TNF processing on the course of colitis in recombination activating gene (RAG)2 -/- mice on transfer of CD4 CD45RB hi T cells. METHODS TNF -/- mice, transgenic for a noncleavable mutant TNF gene, were used as donors of CD4 T cells, and, on a RAG2 -/- background, also as recipients. Kinetics of disease development were compared in the absence of TNF, in the absence of secreted TNF, and in the presence of secreted and tmTNF. The analysis at the end of the observation period included the histopathologic assessment of the intestine and the localization of TNF and interferon gamma (IFNgamma)-expressing cells. RESULTS The complete prevention of TNF secretion in tmTNF transgenic RAG2 -/- mice neither prevented nor delayed disease induction by transferred transgenic for a noncleavable transmembrane mutant of mouse TNF (tmTNF tg) CD4 CD45RB hi T cells. tmTNF expression by transferred CD4 T cells, however, was not required for disease induction because severe colitis and weight loss also were observed in tmTNF RAG2 -/- recipients of TNF -/- CD4 CD45RB hi T cells. In the presence of tmTNF, the absence of secreted TNF did not affect frequency and distribution of TNF and interferon-gamma messenger RNA (mRNA)-expressing cells. CONCLUSIONS These results indicate that specific inhibitors of TNF processing are not appropriate for modulating the pro-inflammatory and disease-inducing effects of TNF in chronic inflammatory disorders of the intestine.
Collapse
Affiliation(s)
- Nadia Corazza
- Department of Pathology, University of Bern, Bern, Switzerland
| | | | | | | | | | | | | |
Collapse
|
23
|
Qi CJ, Zheng L, Zhou X, Tao Y, Ge Y, Zhuang YM, Xu Y, Yu G, Zhang XG. Cross-linking of CD40 using anti-CD40 antibody, 5C11, has different effects on XG2 multiple myeloma cells. Immunol Lett 2004; 93:151-8. [PMID: 15158611 DOI: 10.1016/j.imlet.2004.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2003] [Revised: 02/17/2004] [Accepted: 03/09/2004] [Indexed: 11/30/2022]
Abstract
A multiple myeloma (MM) cell line, XG2, has high-level expression of CD40, a tumor necrosis factor receptor (TNFR) family member. CD40 is present on the surfaces of a large variety of cells, including B cells, endothelial cells, dendritic cells and some carcinoma cells, and delivers signals regulating diverse cellular responses, such as proliferation, differentiation, growth suppression, cell death. In this research, we study the effects of cross-linking of CD40 on myeloma cells using different concentrations of anti-CD40 monoclonal antibody (mAb), 5C11. We found that low concentrations of 5C11 induced proliferation of XG2, while high concentrations of 5C11 resulted in homotypic aggregation of XG2, and strongly suppression of its proliferation and apoptosis after 24 h of treatment. These dose-dependent effects of 5C11 were verified by flow cytometry, Western blotting and immunoprecipitation. Autocrine or paracrine induction of IL-6, and up-regulation of membrane TNF and phosphorylation of TNFR1 may partially explain the contradictory biological effects of CD40 cross-linking on XG2 by anti-CD40 mAb.
Collapse
MESH Headings
- Antibodies, Monoclonal/pharmacology
- Apoptosis/drug effects
- Blotting, Western
- CD40 Antigens/chemistry
- CD40 Antigens/immunology
- CD40 Antigens/metabolism
- Caspase Inhibitors
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cross-Linking Reagents/pharmacology
- Flow Cytometry
- Humans
- Immunoprecipitation
- Interleukin-6/metabolism
- Models, Biological
- Multiple Myeloma/metabolism
- Multiple Myeloma/pathology
- Phosphorylation
- Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type I/immunology
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Tumor Necrosis Factor-alpha/analysis
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Chun-Jian Qi
- Jiangsu Key Laboratory of Clinical Immunology, Institute of Biotechnology, Soochow University, 48 Renmin Road, SuZhou 215007, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kirkegaard T, Pedersen G, Saermark T, Brynskov J. Tumour necrosis factor-alpha converting enzyme (TACE) activity in human colonic epithelial cells. Clin Exp Immunol 2004; 135:146-53. [PMID: 14678276 PMCID: PMC1808921 DOI: 10.1111/j.1365-2249.2004.02348.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Tumour necrosis factor (TNF)-alpha converting enzyme (TACE) releases biologically active, soluble TNF-alpha from transmembrane pro-TNF-alpha and has attracted interest as a specific therapeutic target in inflammatory bowel disease (IBD). Strong immunoreactivity for TACE protein was demonstrated recently in human colonic epithelium, but the function is unknown. We investigated if human colonic epithelial cells express functional TACE activity and how TACE expression is regulated in response to cytokine stimulation. TACE and TNF-alpha mRNA and protein expression were measured in HT-29 and DLD-1 colonic epithelial cells by reverse-transcription polymerase chain reaction, western blotting or enzyme-linked immunosorbent assay. Monocytic THP-1 cells served as positive control. Functional TACE activity was identified and quantified in detergent extracts of cell lines and freshly isolated colonocytes from 14 IBD patients and five controls by a hydrolysis assay using an oligopeptide spanning the cleavage site in pro-TNF-alpha. HT-29 and DLD-1 cells spontaneously expressed TACE mRNA and the active form of TACE protein at levels similar to those of monocytic cells. Functional TACE activity was demonstrated in all cell lines and in cells of controls or IBD patients irrespective of disease activity. TACE mRNA expression and functional activity remained unchanged in cell lines after stimulation with TNF-alpha despite clear induction of TNF-alpha mRNA expression and release of soluble TNF-alpha protein. The release of soluble TNF-alpha protein was almost completely abolished by CH4474, a synthetic TACE inhibitor. We conclude that functional TACE activity is constitutively expressed in human colonic epithelial cells and responsible for processing of the mature, soluble form of TNF-alpha in response to cytokine stimulation.
Collapse
Affiliation(s)
- T Kirkegaard
- Department of Medical Gastroenterology C, Laboratory of Gastroenterology 54O3, Herlev University Hospital, 75 Herlev Ringvej, DK-2730 Herlev, Copenhagen, Denmark
| | | | | | | |
Collapse
|
25
|
Zhang Y, Xu J, Levin J, Hegen M, Li G, Robertshaw H, Brennan F, Cummons T, Clarke D, Vansell N, Nickerson-Nutter C, Barone D, Mohler K, Black R, Skotnicki J, Gibbons J, Feldmann M, Frost P, Larsen G, Lin LL. Identification and Characterization of 4-[[4-(2-Butynyloxy)phenyl]sulfonyl]-N-hydroxy-2,2-dimethyl-(3S)thiomorpholinecarboxamide (TMI-1), a Novel Dual Tumor Necrosis Factor-α-Converting Enzyme/Matrix Metalloprotease Inhibitor for the Treatment of Rheumatoid Arthritis. J Pharmacol Exp Ther 2004; 309:348-55. [PMID: 14718605 DOI: 10.1124/jpet.103.059675] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tumor necrosis factor (TNF)-alpha is a well validated therapeutic target for the treatment of rheumatoid arthritis. TNF-alpha is initially synthesized as a 26-kDa membrane-bound form (pro-TNF) that is cleaved by a Zn-metalloprotease named TNF-alpha-converting enzyme (TACE) to generate the 17-kDa, soluble, mature TNF-alpha. TACE inhibitors that prevent the secretion of soluble TNF-alpha may be effective in treating rheumatoid arthritis (RA) patients. Using a structure-based design approach, we have identified a novel dual TACE/matrix metalloprotease (MMP) inhibitor 4-[[4-(2-butynyloxy)phenyl]sulfonyl]-N-hydroxy-2,2-dimethyl-(3S)thiomorpholinecarboxamide (TMI-1). This molecule inhibits TACE and several MMPs with nanomolar IC(50) values in vitro. In cell-based assays such as monocyte cell lines, human primary monocytes, and human whole blood, it inhibits lipopolysaccharide (LPS)-induced TNF-alpha secretion at submicromolar concentrations, whereas there is no effect on the TNF-alpha mRNA level as judged by RNase protection assay. The inhibition of LPS-induced TNF-alpha secretion is selective because TMI-1 has no effect on the secretion of other proinflammatory cytokines such as interleukin (IL)-1beta, IL-6, and IL-8. Importantly, TMI-1 potently inhibits TNF-alpha secretion by human synovium tissue explants of RA patients. In vivo, TMI-1 is highly effective in reducing clinical severity scores in mouse prophylactic collagen-induced arthritis (CIA) at 5, 10, and 20 mg/kg p.o. b.i.d. and therapeutic CIA model at 100 mg/kg p.o. b.i.d. In summary, TMI-1, a dual TACE/MMP inhibitor, represents a unique class of orally bioavailable small molecule TNF inhibitors that may be effective and beneficial for treating RA.
Collapse
Affiliation(s)
- Yuhua Zhang
- Wyeth Research, 200 Cambridge Park Drive, Cambridge, MA 02140, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Becherer JD, Blobel CP. Biochemical properties and functions of membrane-anchored metalloprotease-disintegrin proteins (ADAMs). Curr Top Dev Biol 2003; 54:101-23. [PMID: 12696747 DOI: 10.1016/s0070-2153(03)54006-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- J David Becherer
- Department of Biochemical and Analytical Pharmacology, GlaxoSmithKline Research Inc., Research Triangle Park, North Carolina 27709, USA
| | | |
Collapse
|
27
|
Beck G, Bottomley G, Bradshaw D, Brewster M, Broadhurst M, Devos R, Hill C, Johnson W, Kim HJ, Kirtland S, Kneer J, Lad N, Mackenzie R, Martin R, Nixon J, Price G, Rodwell A, Rose F, Tang JP, Walter DS, Wilson K, Worth E. (E)-2(R)-[1(S)-(Hydroxycarbamoyl)-4-phenyl-3-butenyl]-2'-isobutyl-2'-(methanesulfonyl)-4-methylvalerohydrazide (Ro 32-7315), a selective and orally active inhibitor of tumor necrosis factor-alpha convertase. J Pharmacol Exp Ther 2002; 302:390-6. [PMID: 12065742 DOI: 10.1124/jpet.302.1.390] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-alpha), a cytokine secreted by inflammatory cells, has been implicated in several inflammatory disease states. (E)-2(R)-[1(S)-(Hydroxycarbamoyl)-4-phenyl-3-butenyl]-2'-isobutyl-2'-(methanesulfonyl)-4-methylvalerohydrazide (Ro 32-7315), is a potent, orally active inhibitor of the TNF-alpha convertase (TACE), an enzyme responsible for proteolytic cleavage of the membrane bound precursor, pro-TNF-alpha. Ro 32-7315 inhibited a recombinant form of TACE (IC(50) = 5.2 nM) with selectivity over related matrix metalloproteinases. In a cellular assay system, THP-1 cell line, and in human and rat whole blood, Ro 32-7315 significantly reduced lipopolysaccharide (LPS)-induced TNF-alpha release with IC(50) values of 350 +/- 14 nM (n = 5), 2.4 +/- 0.5 microM (n = 5), and 110 +/- 18 nM (n = 5), respectively. Oral administration of Ro 32-7315 to Wistar rats caused a dose-dependent inhibition of LPS-induced release of systemic TNF-alpha with an ED(50) of 25 mg/kg. Treatment (days 0-14) of Allen and Hamburys hooded rats with Ro 32-7315 (2.5, 5, 10, and 20 mg/kg, i.p., twice daily) significantly reduced adjuvant-induced secondary paw swelling (42, 71, 83, and 93%, respectively) as compared with the vehicle group. In the Ro 32-7315-treated group, the reduced paw swelling was associated with improved lesion score and joint mobility. Furthermore, in a placebo-controlled, single-dose study, Ro 32-7315 given orally (450 mg) significantly suppressed ex vivo, LPS-induced TNF-alpha release in the whole-blood samples taken from healthy male and female volunteers (mean inhibition of 42% over a 4-h duration, n = 6). These data collectively support the potential use of such a compound for the oral treatment of inflammatory disorders.
Collapse
Affiliation(s)
- G Beck
- Product Lab, Roche Discovery Welwyn, 40 Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AY, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Olleros ML, Guler R, Corazza N, Vesin D, Eugster HP, Marchal G, Chavarot P, Mueller C, Garcia I. Transmembrane TNF induces an efficient cell-mediated immunity and resistance to Mycobacterium bovis bacillus Calmette-Guérin infection in the absence of secreted TNF and lymphotoxin-alpha. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:3394-401. [PMID: 11907097 DOI: 10.4049/jimmunol.168.7.3394] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The contribution of a transmembrane (Tm) form of TNF to protective immunity against Mycobacterium bovis bacillus Calmette-Guérin (BCG) was studied in transgenic (tg) mice expressing a noncleavable Tm TNF but lacking the TNF/lymphotoxin-alpha (LT-alpha) locus (Tm TNF tg mice). These mice were as resistant to BCG infection as wild-type mice, whereas TNF/LT-alpha(-/-), TNF(-/-), and LT-alpha(-/-) mice succumbed. Tm TNF tg mice developed granulomas of smaller size but at 2- to 4-fold increased frequencies compared with wild-type mice. Granulomas were mainly formed by monocytes and activated macrophages expressing Tm TNF mRNA and accumulating acid phosphatase. NO synthase 2 activation as a key macrophage bactericidal mechanism was low during the acute phase of infection in Tm TNF tg mice but was still sufficient to limit bacterial growth and increased in late infection. While infection with virulent Mycobacterium tuberculosis resulted in very rapid death of TNF/LT-alpha(-/-) mice, it also resulted in survival of Tm TNF tg mice which presented an increase in the number of CFU in spleen (5-fold) and lungs (10-fold) as compared with bacterial load of wild-type mice. In conclusion, the Tm form of TNF induces an efficient cell-mediated immunity and total resistance against BCG even in the absence of LT-alpha and secreted TNF. However, Tm TNF-mediated protection against virulent M. tuberculosis infection can also be efficient but not as strong as in BCG infection, in which cognate cellular interactions may play a more predominant role in providing long-term surveillance and containment of BCG-infected macrophages.
Collapse
Affiliation(s)
- Maria L Olleros
- Department of Pathology, University of Geneva, Geneva, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Trifilieff A, Walker C, Keller T, Kottirsch G, Neumann U. Pharmacological profile of PKF242-484 and PKF241-466, novel dual inhibitors of TNF-alpha converting enzyme and matrix metalloproteinases, in models of airway inflammation. Br J Pharmacol 2002; 135:1655-64. [PMID: 11934805 PMCID: PMC1573282 DOI: 10.1038/sj.bjp.0704616] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. TNF-alpha converting enzyme (TACE) and matrix metalloproteinases (MMPs) are believed to play a role in various airway inflammatory disorders. Therefore we have tested the effect of two new inhibitors of TACE/MMPs (PKF242-484, PKF241-466) in models of airway inflammation. 2. PKF242-484 and PKF241-466 inhibited purified MMP-1, -2, -3, -9, -13 and rat collagenase at low nanomolar range. Both compounds inhibited the TNF-alpha release from activated human peripheral blood mononuclear cells with IC(50) values of 56+/-28 and 141+/-100 nM, respectively and had no significant effect on the activation of other human leukocytes, as neither neutrophils and eosinophils oxidative burst nor proliferation or cytokines production by T cells were inhibited in vitro. 3. PKF242-484 and PKF241-466 had beneficial effects in two different murine models of acute lung inflammation in vivo. The influx of neutrophils and lymphocytes into the airways was reduced 3 and 24 h after intranasal LPS challenge. This was accompanied by reduced levels of myeloperoxidase and elastase activities in the bronchoalveolar lavage. Furthermore, a complete inhibition of TNF-alpha release into the airways was observed. In addition, PKF242-484 effectively reduced the influx of neutrophils, eosinophils and lymphocytes in a model of acute allergic lung inflammation. 4. PKF242-484 and PKF241-466 are two novel and potent dual inhibitors of TACE and MMPs, which show activity in in vivo models of lung inflammation. Such compounds could have beneficial effects in airway inflammatory conditions such as asthma and chronic obstructive pulmonary disease.
Collapse
|
30
|
Abstract
Leukocyte extravasation into tissues is a multi-step process culminating in the migration of cells through the basement membrane. This requires the production of matrix-degrading enzymes, in particular matrix metalloproteinases (MMP). We investigated the role of chemokines in regulating MMP production in the monocytic cell line THP-1 and in peripheral blood monocytes (PBM). The CC chemokines CCL2 (MCP-1), CCL3 (MIP-1alpha), and CCL5 (RANTES) stimulated the release of monocyte MMP-9 protein in a bell-shaped dose-dependent manner. The increase in MMP-9 protein detected at 24 h was due to de novo synthesis, confirmed by Northern blotting, with MMP-9 mRNA detectable at 6-8 h. Autocrine TNF-alpha was necessary for chemokine stimulation of MMP-9. Chemokines increased TNF-alpha mRNA levels and protein release in monocytes and THP-1 cells, and neutralizing anti-TNF-alpha antibodies inhibited CCL2-induced MMP-9 release. Furthermore, the broad spectrum MMP inhibitor BB 2516, which inhibits TNF-alpha release, abrogated CCL2- and CCL5-induced MMP-9 release in both THP-1 cells and freshly isolated monocytes. Monocyte production of MMP is of major importance in the pathology of cancer, asthma, and rheumatoid arthritis. An understanding of the mechanisms by which these MMP are produced may lead to novel therapies to modulate extravasation of leukocytes in disease.
Collapse
Affiliation(s)
- Stephen C Robinson
- ICRF Translational Oncology Laboratory, Bart's and the London Queen Mary School of Dentistry, Charterhouse Square, London, EC1M 6BQ, GB.
| | | | | |
Collapse
|
31
|
|
32
|
Affiliation(s)
- K A Papadakis
- Division of Gastroenterology and Inflammatory Bowel Disease Center, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, California 90048, USA
| | | |
Collapse
|
33
|
Dekkers PE, ten Hove T, Lauw FN, Koene HR, Lumley P, van Deventer SJ, van der Poll T. The metalloproteinase inhibitor GI5402 inhibits endotoxin-induced soluble CD27 and CD16 release in healthy humans. Infect Immun 2000; 68:3036-9. [PMID: 10769011 PMCID: PMC97526 DOI: 10.1128/iai.68.5.3036-3039.2000] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Metalloproteinases have been implicated in the cleavage of a number of cell surface immune receptors. Oral administration of the metalloproteinase inhibitor GI5402 attenuated the release of soluble CD27 and CD16 into the circulation after intravenous endotoxin injection in healthy humans.
Collapse
Affiliation(s)
- P E Dekkers
- Laboratory of Experimental Internal Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|