1
|
Opperman CJ, Wojno J, Goosen W, Warren R. Phages for the treatment of Mycobacterium species. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 201:41-92. [PMID: 37770176 DOI: 10.1016/bs.pmbts.2023.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Highly drug-resistant strains are not uncommon among the Mycobacterium genus, with patients requiring lengthy antibiotic treatment regimens with multiple drugs and harmful side effects. This alarming increase in antibiotic resistance globally has renewed the interest in mycobacteriophage therapy for both Mycobacterium tuberculosis complex and non-tuberculosis mycobacteria. With the increasing number of genetically well-characterized mycobacteriophages and robust engineering tools to convert temperate phages to obligate lytic phages, the phage cache against extensive drug-resistant mycobacteria is constantly expanding. Synergistic effects between phages and TB drugs are also a promising avenue to research, with mycobacteriophages having several additional advantages compared to traditional antibiotics due to their different modes of action. These advantages include less side effects, a narrow host spectrum, biofilm penetration, self-replication at the site of infection and the potential to be manufactured on a large scale. In addition, mycobacteriophage enzymes, not yet in clinical use, warrant further studies with their additional benefits for rupturing host bacteria thereby limiting resistance development as well as showing promise in vitro to act synergistically with TB drugs. Before mycobacteriophage therapy can be envisioned as part of routine care, several obstacles must be overcome to translate in vitro work into clinical practice. Strategies to target intracellular bacteria and selecting phage cocktails to limit cross-resistance remain important avenues to explore. However, insight into pathophysiological host-phage interactions on a molecular level and innovative solutions to transcend mycobacteriophage therapy impediments, offer sufficient encouragement to explore phage therapy. Recently, the first successful clinical studies were performed using a mycobacteriophage-constructed cocktail to treat non-tuberculosis mycobacteria, providing substantial insight into lessons learned and potential pitfalls to avoid in order to ensure favorable outcomes. However, due to mycobacterium strain variation, mycobacteriophage therapy remains personalized, only being utilized in compassionate care cases until there is further regulatory approval. Therefore, identifying the determinants that influence clinical outcomes that can expand the repertoire of mycobacteriophages for therapeutic benefit, remains key for their future application.
Collapse
Affiliation(s)
- Christoffel Johannes Opperman
- National Health Laboratory Service, Green Point TB-Laboratory, Cape Town, South Africa; DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for tuberculosis Research, Division of Molecular Biology and Human Genetics, Stellenbosch University, Cape Town, South Africa; Division of Medical Microbiology, University of Cape Town, Cape Town, South Africa.
| | - Justyna Wojno
- Division of Medical Microbiology, University of Cape Town, Cape Town, South Africa; Lancet Laboratories, Cape Town, South Africa
| | - Wynand Goosen
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for tuberculosis Research, Division of Molecular Biology and Human Genetics, Stellenbosch University, Cape Town, South Africa
| | - Rob Warren
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, SAMRC Centre for tuberculosis Research, Division of Molecular Biology and Human Genetics, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
2
|
Haferland I, Wallenwein CM, Ickelsheimer T, Diehl S, Wacker MG, Schiffmann S, Buerger C, Kaufmann R, Koenig A, Pinter A. Mechanism of anti-inflammatory effects of Rifampicin in an ex vivo culture system of Hidradenitis Suppurativa. Exp Dermatol 2022; 31:1005-1013. [PMID: 35048417 DOI: 10.1111/exd.14531] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 11/30/2022]
Abstract
Hidradenitis suppurativa (HS) is a chronic inflammatory skin disease of the hair follicles leading to painful lesions, associated with increased levels of pro-inflammatory cytokines. Numerous guidelines recommend antibiotics like clindamycin and rifampicin in combination, as first-line systemic therapy in moderate to severe forms of inflammation. HS has been proposed to be mainly an auto-inflammatory disease associated with but not initially provoked by bacteria. Therefore, it has to be assumed that the pro-inflammatory milieu previously observed in HS skin is not solely dampened by the bacteriostatic inhibition of DNA-dependent RNA polymerase. To further clarify the mechanism of anti-inflammatory effects of rifampicin, ex vivo explants of lesional HS from 8 HS patients were treated with rifampicin, and its effect on cytokine production, immune cells as well as the expression of Toll-like receptor 2 (TLR2) were investigated. Analysis of cell culture medium of rifampicin treated HS explants revealed an anti-inflammatory effect of rifampicin that significantly inhibiting interleukin (IL)-1β, IL-6, IL-8, IL-10, and tumour necrosis factor (TNF) -α production. Immunohistochemistry of the rifampicin-treated explants suggested a tendency for it to reduce the expression of TLR2 while not affecting the number of immune cells.
Collapse
Affiliation(s)
- Isabel Haferland
- Department of Dermatology, Venerology and Allergology, University Hospital Frankfurt am Main, Frankfurt am Main, Germany
| | - Chantal M Wallenwein
- Fraunhofer Insitute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Tanja Ickelsheimer
- Department of Dermatology, Venerology and Allergology, University Hospital Frankfurt am Main, Frankfurt am Main, Germany
| | - Sandra Diehl
- Department of Dermatology, Venerology and Allergology, University Hospital Frankfurt am Main, Frankfurt am Main, Germany
| | - Matthias G Wacker
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Susanne Schiffmann
- Fraunhofer Insitute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Claudia Buerger
- Department of Dermatology, Venerology and Allergology, University Hospital Frankfurt am Main, Frankfurt am Main, Germany
| | - Roland Kaufmann
- Department of Dermatology, Venerology and Allergology, University Hospital Frankfurt am Main, Frankfurt am Main, Germany
| | - Anke Koenig
- Department of Dermatology, Venerology and Allergology, University Hospital Frankfurt am Main, Frankfurt am Main, Germany
| | - Andreas Pinter
- Department of Dermatology, Venerology and Allergology, University Hospital Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
3
|
SARS-CoV-2 RdRp Inhibitors Selected from a Cell-Based SARS-CoV-2 RdRp Activity Assay System. Biomedicines 2021; 9:biomedicines9080996. [PMID: 34440200 PMCID: PMC8392292 DOI: 10.3390/biomedicines9080996] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/03/2021] [Accepted: 08/10/2021] [Indexed: 01/18/2023] Open
Abstract
The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), urgently needs effective prophylactic and therapeutic drugs. RNA-dependent RNA polymerase (RdRp), essential for replicating and transcribing a viral RNA genome, is highly conserved in coronaviruses; thus, it is a potential target for inhibiting coronavirus infection. In this study, we generated the cell-based SARS-CoV-2 RdRp activity assay system by modifying a previously reported cell-based MERS-CoV RdRp activity assay system to screen for SARS-CoV-2 RdRp inhibitors. The assay system consisted of an expression plasmid encoding SARS-CoV-2 RdRp and an RdRp activity reporter plasmid. RdRp activity in the cells could be conveniently detected by luminescence after transfection. We confirmed that SARS-CoV-2 RdRp replicated double-stranded RNA using immunofluorescence staining and the inhibition of RdRp activity by remdesivir and lycorine using this system. Moreover, the Z-factor of this system was calculated to be 0.798, suggesting the reproducibility and reliability of the high-throughput screening system. Finally, we screened nucleoside and nucleotide analogs and identified adefovir dipivoxil, emtricitabine, telbivudine, entecavir hydrate, moroxydine and rifampin as novel SARS-CoV-2 RdRp inhibitors and therapeutic candidates for COVID-19 This system provides an effective high-throughput screening system platform for developing potential prophylactic and therapeutic drugs for COVID-19 and emerging coronavirus infections.
Collapse
|
4
|
The Effects of Rifampicin on Experimental Cerebral Ischemia/Reperfusion Injury in Rats. IRANIAN RED CRESCENT MEDICAL JOURNAL 2019. [DOI: 10.5812/ircmj.87869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
5
|
Tükenmez H, Edström I, Kalsum S, Braian C, Ummanni R, Fick SB, Sundin C, Lerm M, Elofsson M, Larsson C. Corticosteroids protect infected cells against mycobacterial killing in vitro. Biochem Biophys Res Commun 2019; 511:117-121. [PMID: 30773257 DOI: 10.1016/j.bbrc.2019.02.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 02/08/2019] [Indexed: 12/23/2022]
Abstract
The effect of corticosteroids on human physiology is complex and their use in tuberculosis patients remains controversial. In a high-throughput screening approach designed to discover virulence inhibitors, several corticosteroids were found to prevent cytolysis of fibroblasts infected with mycobacteria. Further experiments with Mycobacterium tuberculosis showed anti-cytolytic activity in the 10 nM range, but no effect on bacterial growth or survival in the absence of host cells at 20 μM. The results from a panel of corticosteroids with various affinities to the glucocorticoid- and mineralocorticoid receptors indicate that the inhibition of cytolysis most likely is mediated through the glucocorticoid receptor. Using live-imaging of M. tuberculosis-infected human monocyte-derived macrophages, we also show that corticosteroids to some extent control intracellular bacteria. In vitro systems with reduced complexity are to further study and understand the interactions between bacterial infection, immune defense and cell signaling.
Collapse
Affiliation(s)
- Hasan Tükenmez
- Department of Molecular Biology, Umeå University, SE-901 87, Umeå, Sweden
| | - Isabel Edström
- Department of Molecular Biology, Umeå University, SE-901 87, Umeå, Sweden
| | - Sadaf Kalsum
- Department of Clinical and Experimental Medicine, Linköping University, SE-581 83, Linköping, Sweden
| | - Clara Braian
- Department of Clinical and Experimental Medicine, Linköping University, SE-581 83, Linköping, Sweden
| | - Ramesh Ummanni
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Hyderabad, 500007, Telangana, India
| | - Stina Berglund Fick
- Chemical Biology Consortium Sweden (CBCS), Department of Chemistry, Umeå University, SE-901 87, Umeå, Sweden
| | - Charlotta Sundin
- Department of Chemistry, Umeå University, SE-901 87, Umeå, Sweden
| | - Maria Lerm
- Department of Clinical and Experimental Medicine, Linköping University, SE-581 83, Linköping, Sweden
| | - Mikael Elofsson
- Department of Chemistry, Umeå University, SE-901 87, Umeå, Sweden
| | - Christer Larsson
- Infectious Diseases Clinic, Umeå University Hospital, SE-901 85, Umeå, Sweden.
| |
Collapse
|
6
|
Zahednasab H, Firouzi M, Kaboudanian-Ardestani S, Mojallal-Tabatabaei Z, Karampour S, Keyvani H. The protective effect of rifampicin on behavioral deficits, biochemical, and neuropathological changes in a cuprizone model of demyelination. Cytokine 2019; 113:417-426. [DOI: 10.1016/j.cyto.2018.10.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/16/2018] [Accepted: 10/18/2018] [Indexed: 12/19/2022]
|
7
|
López-Ramos M, Figueroa-Valverde L, Herrera-Meza S, Rosas-Nexticapa M, Díaz-Cedillo F, García-Cervera E, Pool-Gómez E, Cahuich-Carrillo R. Design and synthesis of a new steroid-macrocyclic derivative with biological activity. J Chem Biol 2017; 10:69-84. [PMID: 28405241 PMCID: PMC5374095 DOI: 10.1007/s12154-017-0165-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 02/02/2017] [Indexed: 10/20/2022] Open
Abstract
The aims of this study were to evaluate the positive inotropic effect of a new macrocyclic derivative (compound 11) and characterize the molecular mechanism involved in its biological activity. The first step was achieved by synthesis of a macrocyclic derivative involving a series of reactions for the preparation of several steroid derivatives such as (a) steroid-pyrimidinone (3 and 4), (b) steroid-amino (5), (c) steroid-imino (6), (d) ester-steroid (7 and 8), and (e) amido-steroid (9 and 10). Finally, 11 was prepared by removing the tert-butyldimethylsilane fragment of 10. The biological activity of compounds on perfusion pressure and vascular resistance was evaluated on isolated rat heart using the Langendorff model. The inotropic activity of 11 was evaluated in presence of prazosin, metoprolol, indomethacin, nifedipine, and flutamide to characterize its molecular mechanism. Theoretical experiments were carried out with a Docking model, to assess potential interactions of androgen receptor with 11. The results showed that only this macrocyclic derivative exerts changes on perfusion pressure and vascular resistance translated as the positive inotropic effect, and this effect was blocked with flutamide; these data indicate that the positive inotropic activity induced by this macrocyclic derivative was via androgen receptor activation. The theoretical results indicated that the interaction of the macrocyclic derivative with the androgen receptor involves several amino acid residues such as Leu704, Asn705, Met780, Cys784, Met749, Leu762, Phe764, Ser778, and Met787. In conclusion, all these data suggest that the positive inotropic activity of the macrocyclic derivative may depend on its chemical structure.
Collapse
Affiliation(s)
- Maria López-Ramos
- Laboratory of Pharmacochemistry, University Autonomous of Campeche, Av. Agustín Melgar s/n, Col Buenavista, C.P. 24039 Campeche, Mexico
| | - Lauro Figueroa-Valverde
- Laboratory of Pharmacochemistry, University Autonomous of Campeche, Av. Agustín Melgar s/n, Col Buenavista, C.P. 24039 Campeche, Mexico
| | - Socorro Herrera-Meza
- Instituto de Investigaciones Psicológicas, Universidad Veracruzana, Av. Dr. Luis Castelazo Ayala s/n Col Industrial Animas, C.P. 91190 Xalapa, VER Mexico
| | - Marcela Rosas-Nexticapa
- Facultad de Nutrición, Universidad Veracruzana, Médicos y Odontologos s/n, Unidad del Bosque, C.P. 91010 Xalapa, VER Mexico
| | - Francisco Díaz-Cedillo
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. Carpio y Plan de Ayala s/n Col, C.P. 11340 Santo Tomas, DF Mexico
| | - Elodia García-Cervera
- Laboratory of Pharmacochemistry, University Autonomous of Campeche, Av. Agustín Melgar s/n, Col Buenavista, C.P. 24039 Campeche, Mexico
| | - Eduardo Pool-Gómez
- Laboratory of Pharmacochemistry, University Autonomous of Campeche, Av. Agustín Melgar s/n, Col Buenavista, C.P. 24039 Campeche, Mexico
| | - Regina Cahuich-Carrillo
- Laboratory of Pharmacochemistry, University Autonomous of Campeche, Av. Agustín Melgar s/n, Col Buenavista, C.P. 24039 Campeche, Mexico
| |
Collapse
|
8
|
Ivashchenko O, Coy E, Peplinska B, Jarek M, Lewandowski M, Załęski K, Warowicka A, Wozniak A, Babutina T, Jurga-Stopa J, Dolinsek J, Jurga S. Influence of silver content on rifampicin adsorptivity for magnetite/Ag/rifampicin nanoparticles. NANOTECHNOLOGY 2017; 28:055603. [PMID: 28029097 DOI: 10.1088/1361-6528/28/5/055603] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Magnetite nanoparticles (NPs) decorated with silver (magnetite/Ag) are intensively investigated due to their application in the biomedical field. We demonstrate that the increase of silver content on the surface of nanoparticles improves the adsorptivity of antibiotic rifampicin as well as antibacterial properties. The use of ginger extract allowed to improve the silver nucleation on the magnetite surface that resulted in an increase of silver content. Physicochemical and functional characterization of magnetite/Ag NPs was performed. Our results show that 5%-10% of silver content in magnetite/Ag NPs is already sufficient for antimicrobial properties against Streptococcus salivarius and Staphylococcus aureus. The rifampicin molecules on the magnetite/Ag NPs surface made the spectrum of antimicrobial activity wider. Cytotoxicity evaluation of the magnetite/Ag/rifampicin NPs showed no harmful action towards normal human fibroblasts, whereas the effect on human embryonic kidney cell viability was time and dose dependent.
Collapse
|
9
|
Ivashchenko O, Woźniak A, Coy E, Peplinska B, Gapinski J, Jurga S. Release and cytotoxicity studies of magnetite/Ag/antibiotic nanoparticles: An interdependent relationship. Colloids Surf B Biointerfaces 2017; 152:85-94. [PMID: 28088016 DOI: 10.1016/j.colsurfb.2017.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 01/05/2017] [Accepted: 01/06/2017] [Indexed: 01/20/2023]
Abstract
Though the cytotoxic properties of magnetite nanoparticles (NPs) are rather well investigated and known to be dose dependent and rather low, surface functionalization can drastically change their properties. To determine whether the cytotoxicity of magnetite/Ag/antibiotic NPs may be associated, among other things, with iron, silver and antibiotic release, this study investigates the release profiles and cytotoxicity of magnetite/Ag/rifampicin and magnetite/Ag/doxycycline NPs compares it similar profiles from magnetite, magnetite/Ag NPs and antibiotics. It was established that the studied NPs released not only water-soluble substances, such as antibiotics, but also poorly-soluble ones, such as iron and silver. The deposition of silver on the magnetite surface promotes the release of iron by the formation of a galvanic couple. Antibiotic adsorbed on the magnetite/Ag surface plays a dual role in the galvanic corrosion processes: as a corrosion inhibitor for iron oxides and as a corrosion promoter for silver. Magnetite/Ag/rifampicin and magnetite/Ag/doxycycline. NPs were found to have greater cytotoxicity towards the HEK293T cell line than magnetite NPs. These results were attributed to the combined toxic action of the released iron, silver ions and antibiotics. Intensive and simultaneous release of the NP components caused cell stress and suppressed their growth.
Collapse
Affiliation(s)
- Olena Ivashchenko
- NanoBioMedical Centre, Adam Mickiewicz University, 61614 Poznan, Poland.
| | - Anna Woźniak
- NanoBioMedical Centre, Adam Mickiewicz University, 61614 Poznan, Poland
| | - Emerson Coy
- NanoBioMedical Centre, Adam Mickiewicz University, 61614 Poznan, Poland
| | - Barbara Peplinska
- NanoBioMedical Centre, Adam Mickiewicz University, 61614 Poznan, Poland
| | - Jacek Gapinski
- Department of Molecular Biophysics, Faculty of Physics, A. Mickiewicz University, Poznań, Poland
| | - Stefan Jurga
- NanoBioMedical Centre, Adam Mickiewicz University, 61614 Poznan, Poland; Department of Macromolecular Physics, Adam Mickiewicz University, 61614 Poznan, Poland
| |
Collapse
|
10
|
Shehu AI, Li G, Xie W, Ma X. The pregnane X receptor in tuberculosis therapeutics. Expert Opin Drug Metab Toxicol 2015; 12:21-30. [PMID: 26592418 DOI: 10.1517/17425255.2016.1121381] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Among the infectious diseases, tuberculosis (TB) remains the second most common cause of death after HIV. TB treatment requires the combination of multiple drugs including the rifamycin class. However, rifamycins are activators of human pregnane X receptor (PXR), a transcription factor that regulates drug metabolism, drug resistance, energy metabolism and immune response. Rifamycin-mediated PXR activation may affect the outcome of TB therapy. AREAS COVERED This review describes the role of PXR in modulating metabolism, efficacy, toxicity and resistance to anti-TB drugs; as well as polymorphisms of PXR that potentially affect TB susceptibility. EXPERT OPINION The wide range of PXR functions that mediate drug metabolism and toxicity in TB therapy are often underappreciated and thus understudied. Further studies are needed to determine the overall impact of PXR activation on the outcome of TB therapy.
Collapse
Affiliation(s)
- Amina I Shehu
- a Center for Pharmacogenetics, Department of Pharmaceutical Sciences , School of Pharmacy, University of Pittsburgh , Pittsburgh , PA 15261 , USA
| | - Guangming Li
- b Department of Hepatology, the 6th People's Hospital of Zhengzhou , the Hospital for Infectious Diseases in Henan Province , Zhengzhou , China
| | - Wen Xie
- a Center for Pharmacogenetics, Department of Pharmaceutical Sciences , School of Pharmacy, University of Pittsburgh , Pittsburgh , PA 15261 , USA
| | - Xiaochao Ma
- a Center for Pharmacogenetics, Department of Pharmaceutical Sciences , School of Pharmacy, University of Pittsburgh , Pittsburgh , PA 15261 , USA
| |
Collapse
|
11
|
RIFAMPICIN: an antibiotic with brain protective function. Brain Res Bull 2014; 107:37-42. [PMID: 24905548 DOI: 10.1016/j.brainresbull.2014.05.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/08/2014] [Accepted: 05/27/2014] [Indexed: 11/22/2022]
Abstract
Besides its well known antibiotic activity rifampicin exerts multiple brain protective functions in acute cerebral ischemia and chronic neurodegeneration. The present mini-review gives an update of the unique activity of rifampicin in different diseases including Parkinson's disease, meningitis, stroke, Alzheimer's disease and optic nerve injury.
Collapse
|
12
|
Cakici O, Aksak S, Unal D, Sipal S, Keles S, Dumlu T, Karamese M. Effects of intraocular rifampicin on retinal ganglion cell structure: a stereological and histopathological study. Int J Ophthalmol 2013; 6:596-9. [PMID: 24195032 DOI: 10.3980/j.issn.2222-3959.2013.05.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 08/02/2013] [Indexed: 11/02/2022] Open
Abstract
AIM To determine the histopathological changes of rifampicin applied intravitreally on retinal ganglion cells by means of stereological and histopathological methods. METHODS For this study twenty-four New Zealand adult rabbits were divided into four groups (n=6 for each group). 50µg/0.1mL (group 1), 100µg/0.1mL (group 2), 150µg/0.1mL (group 3) and 200µg/0.1mL (group 4), rifampicin were injected into the vitreous of the right eyes of animals, their left eyes were used as control (group 5). After the 28(th) day of application, animals were anesthetised with xylazine (8mg/kg, IM) and then their eyes were enucleated immediately. Patterns were taken away and eyes were prepared for both stereological and electromicroscopic observation. RESULTS Depending on the high dose of rifampicin, some histopathological changes such as cytoplasmic dilatation and damaged membrane were observed on the electromicroscopic level. Using quantitative examination, which was done at the light microscopic level, it was shown that the number of neurons decreased linearly as rifampicin dose increased when compared with the control group. CONCLUSION Based on these findings, low-dose rifampicin (50µg/0.1mL) may be useful for treatment of the ocular diseases.
Collapse
Affiliation(s)
- Ozgür Cakici
- Department of Ophthalmology, Erzurum Research and Education Hospital, Erzurum 25100, Turkey
| | | | | | | | | | | | | |
Collapse
|
13
|
Vancomycin-rifampin combination therapy has enhanced efficacy against an experimental Staphylococcus aureus prosthetic joint infection. Antimicrob Agents Chemother 2013; 57:5080-6. [PMID: 23917317 DOI: 10.1128/aac.00702-13] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Treatment of prosthetic joint infections often involves a two-stage exchange, with implant removal and antibiotic spacer placement followed by systemic antibiotic therapy and delayed reimplantation. However, if antibiotic therapy can be improved, one-stage exchange or implant retention may be more feasible, thereby decreasing morbidity and preserving function. In this study, a mouse model of prosthetic joint infection was used in which Staphylococcus aureus was inoculated into a knee joint containing a surgically placed metallic implant extending from the femur. This model was used to evaluate whether combination therapy of vancomycin plus rifampin has increased efficacy compared with vancomycin alone against these infections. On postoperative day 7, vancomycin with or without rifampin was administered for 6 weeks with implant retention. In vivo bioluminescence imaging, ex vivo CFU enumeration, X-ray imaging, and histologic analysis were carried out. We found that there was a marked therapeutic benefit when vancomycin was combined with rifampin compared with vancomycin alone. Taken together, our results suggest that the mouse model used could serve as a valuable in vivo preclinical model system to evaluate and compare efficacies of antibiotics and combinatory therapy for prosthetic joint infections before more extensive studies are carried out in human subjects.
Collapse
|
14
|
Shear stress activation of nuclear receptor PXR in endothelial detoxification. Proc Natl Acad Sci U S A 2013; 110:13174-9. [PMID: 23878263 DOI: 10.1073/pnas.1312065110] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Endothelial cells (ECs) are constantly exposed to xenobiotics and endobiotics or their metabolites, which perturb EC function, as well as to shear stress, which plays a crucial role in vascular homeostasis. Pregnane X receptor (PXR) is a nuclear receptor and a key regulator of the detoxification of xeno- and endobiotics. Here we show that laminar shear stress (LSS), the atheroprotective flow, activates PXR in ECs, whereas oscillatory shear stress, the atheroprone flow, suppresses PXR. LSS activation of PXR in cultured ECs led to the increased expression of a PXR target gene, multidrug resistance 1 (MDR1). An in vivo study using rats showed that the expression of MDR1 was significantly higher in the endothelium from the descending thoracic aorta, where flow is mostly laminar, than from the inner curvature of aortic arch, where flow is disturbed. Functionally, LSS-activated PXR protects ECs from apoptosis triggered by doxorubicin via the induction of MDR1 and other detoxification genes. PXR also suppressed the expression of proinflammatory adhesion molecules and monocyte adhesion in response to TNF-α and lipopolysaccharide. Overexpression of a constitutively active PXR in rat carotid arteries potently attenuated proinflammatory responses. In addition, cDNA microarray revealed a large number of the PXR-activated endothelial genes whose products are responsible for major steps of detoxification, including phase I and II metabolizing enzymes and transporters. These detoxification genes in ECs are induced by LSS in ECs in a PXR-dependent manner. In conclusion, our results indicate that PXR represents a flow-activated detoxification system to protect ECs against damage by xeno- and endobiotics.
Collapse
|
15
|
Kwiatkowska B, Maslinska M, Przygodzka M, Dmowska-Chalaba J, Dabrowska J, Sikorska-Siudek K. Immune system as a new therapeutic target for antibiotics. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/abb.2013.44a013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
16
|
Abstract
Rifampicin is a macrocyclic antibiotic used extensively for the treatment of Mycobacterium tuberculosis and other mycobacterial infections. Recently, it was discovered that rifampicin exhibits neuroprotective effects. It has been shown to protect PC12 cells against MPP(+)-induced apoptosis and inhibit the expression of α-synuclein multimers. In in vitro studies, rifampicin pretreatment protects PC12 cells against rotenone-induced cell death. Qualitative and quantitative analyses uncover that rifampicin significantly suppresses rotenone-induced apoptosis by ameliorating mitochondrial oxidative stress. It reduces microglial inflammation and improves neuron survival. Our results indicate that rifampicin is cytoprotective under a variety of experimental conditions, and suggest that it may be useful in PD therapeutics. It is the aim of this paper to review the experimental neuroprotection data reported using rifampicin with a focus on the molecular and cellular mechanisms of cytoprotective effect in in vitro models of PD.
Collapse
|
17
|
Bi W, Jing X, Zhu L, Liang Y, Liu J, Yang L, Xiao S, Xu A, Shi Q, Tao E. Inhibition of 26S protease regulatory subunit 7 (MSS1) suppresses neuroinflammation. PLoS One 2012; 7:e36142. [PMID: 22629310 PMCID: PMC3356363 DOI: 10.1371/journal.pone.0036142] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 03/26/2012] [Indexed: 11/18/2022] Open
Abstract
Recently, researchers have focused on immunosuppression induced by rifampicin. Our previous investigation found that rifampicin was neuroprotective by inhibiting the production of pro-inflammatory mediators, thereby suppressing microglial activation. In this study, using 2-dimensional gel electrophoresis (2-DE) and mass spectrometry (MS), we discovered that 26S protease regulatory subunit 7 (MSS1) was decreased in rifampicin-treated microglia. Western blot analysis verified the downregulation of MSS1 expression by rifampicin. As it is indicated that the modulation of the ubiquitin-26S proteasome system (UPS) with proteasome inhibitors is efficacious for the treatment of neuro-inflammatory disorders, we next hypothesized that silencing MSS1 gene expression might inhibit microglial inflammation. Using RNA interference (RNAi), we showed significant reduction of IkBα degradation and NF-kB activation. The production of lipopolysaccharides-induced pro-inflammatory mediators such as inducible nitric oxide synthase (iNOS), nitric oxide, cyclooxygenase-2, and prostaglandin E2 were also reduced by MSS1 gene knockdown. Taken together, our findings suggested that rifampicin inhibited microglial inflammation by suppressing MSS1 protein production. Silencing MSS1 gene expression decreased neuroinflammation. We concluded that MSS1 inhibition, in addition to anti-inflammatory rifampicin, might represent a novel mechanism for the treatment of neuroinflammatory disorders.
Collapse
Affiliation(s)
- Wei Bi
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Neurology, First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Xiuna Jing
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Lihong Zhu
- Joint Laboratory for Brain Function and Health of Jinan University and the University of Hong Kong, School of Medicine, Jinan University, Guangzhou, People’s Republic of China
| | - Yanran Liang
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Jun Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Lianhong Yang
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Songhua Xiao
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Anding Xu
- Department of Neurology, First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Qiaoyun Shi
- Division of Cardiovascular Medicine, Center for Inherited Cardiovascular Disease, Stanford University School of Medicine, Stanford, California, United States of America
| | - Enxiang Tao
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
- * E-mail:
| |
Collapse
|
18
|
Vandevyver S, Dejager L, Libert C. On the trail of the glucocorticoid receptor: into the nucleus and back. Traffic 2011; 13:364-74. [PMID: 21951602 DOI: 10.1111/j.1600-0854.2011.01288.x] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 09/20/2011] [Accepted: 09/20/2011] [Indexed: 01/20/2023]
Abstract
The glucocorticoid receptor (GR) belongs to the superfamily of steroid receptors and is an important regulator of physiological and metabolic processes. In its inactive state, GR is unbound by ligand and resides in the cytoplasm in a chaperone complex. When it binds glucocorticoids, it is activated and translocates to the nucleus, where it functions as a transcription factor. However, the subcellular localization of GR is determined by the balance between its rates of nuclear import and export. The mechanism of GR nuclear transport has been extensively studied. Originally, it was believed that nuclear import of GR is initiated by dissociation of the chaperone complex in the cytoplasm. However, several studies show that the chaperone machinery is required for nuclear transport of GR. In this review, we summarize the contribution of various chaperone components involved in the nuclear transport of GR and propose an updated model of its nuclear import and export. Moreover, we review the importance of ligand-independent nuclear transport and compare the nuclear transport of GR with that of other steroid receptors.
Collapse
Affiliation(s)
- Sofie Vandevyver
- Department of Molecular Biomedical Research, VIB, Ghent University, FSVM Building, Technologiepark 927, B9052, Ghent, Belgium
| | | | | |
Collapse
|
19
|
Bi W, Zhu L, Wang C, Liang Y, Liu J, Shi Q, Tao E. Rifampicin inhibits microglial inflammation and improves neuron survival against inflammation. Brain Res 2011; 1395:12-20. [PMID: 21555117 DOI: 10.1016/j.brainres.2011.04.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 04/08/2011] [Accepted: 04/12/2011] [Indexed: 12/21/2022]
Abstract
Microglial activation plays an important role in the pathophysiology of neurodegenerative diseases, and suppression of microglial activation prevents the progression of neurodegeneration. Rifampicin, a bacteriocidal antibiotic, induces immunosuppression. We hypothesized that rifampicin might be neuroprotective by inhibiting the production of pro-inflammatory mediators, thereby suppressing microglial activation. In the present study, we examined the effects of rifampicin on the production of lipopolysaccharide (LPS)-induced pro-inflammatory mediators and their signaling pathways in BV2 microglia. We also assessed the neuroprotective effects of rifampicin using a co-culture of microglia and neurons. Our results showed that rifampicin inhibited the LPS-stimulated expression of inducible nitric oxide synthase, cyclooxygenase-2, tumor necrosis factor-α, and interleukin-1β, as well as the production of nitric oxide and prostaglandin E₂. Moreover, rifampicin suppressed LPS-induced nuclear factor-kappa B activation by blocking the degradation of the inhibitor of the nuclear transcription factor NF-kappa B. Rifampicin inhibited the phosphorylation of mitogen activated protein kinases, although protein kinase B was not inhibited. Preincubation of microglia with rifampicin reduced neurotoxicity and improved neuron survival in a microglia-neuronal co-culture system. Taken together, these findings suggest that rifampicin, with its anti-inflammatory properties, might be a novel treatment for neurodegenerative diseases.
Collapse
Affiliation(s)
- Wei Bi
- Department of Neurology, Sun Yat-sen memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China
| | | | | | | | | | | | | |
Collapse
|
20
|
Smani Y, Domínguez-Herrera J, Pachón J. Rifampin Protects Human Lung Epithelial Cells Against Cytotoxicity Induced by Clinical Multi and Pandrug-resistant Acinetobacter baumannii. J Infect Dis 2011; 203:1110-9. [DOI: 10.1093/infdis/jiq159] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
21
|
Dey A, Verma AK, Chatterji D. Molecular insights into the mechanism of phenotypic tolerance to rifampicin conferred on mycobacterial RNA polymerase by MsRbpA. MICROBIOLOGY-SGM 2011; 157:2056-2071. [PMID: 21415119 DOI: 10.1099/mic.0.047480-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The protein MsRbpA from Mycobacterium smegmatis rescues RNA polymerase (RNAP) from the inhibitory effect of rifampicin (Rif). We have reported previously that MsRbpA interacts with the β-subunit of RNAP and that the effect of MsRbpA on Rif-resistant (Rif(R)) RNAP is minimal. Here we attempted to gain molecular insights into the mechanism of action of this protein with respect to its role in rescuing RNAP from Rif-mediated transcription inhibition. Our experimental approach comprised multiple-round transcription assays, fluorescence spectroscopy, MS and surface plasmon resonance in order to meet the above objective. Based on our molecular studies we propose here that Rif is released from its binding site in the RNAP-Rif complex in the presence of MsRbpA. Biophysical studies reveal that the location of MsRbpA on RNAP is at the junction of the β- and β'-subunits, close to the Rif-binding site and the (i+1) site on RNAP.
Collapse
Affiliation(s)
- Abhinav Dey
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| | - Amit Kumar Verma
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| | - Dipankar Chatterji
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
22
|
Abstract
Intraocular manifestations of tuberculosis (TB) are rare, but TB infection is common worldwide, especially in developing economies, and in immigrant populations and immunocompromised patients in developed nations. The current review focuses on the clinical characteristics and diagnostic modalities useful in the diagnosis of intraocular TB. Specifically, IFN-gamma Release Assays (IGRAs), antigen-detection assays, and polymerase chain reactions will be discussed. Clinical management of TB patients includes counseling and testing for HIV infection. The use of corticosteroids along with anti-tuberculous medications and special therapeutic considerations in immunocompromised patients are discussed.
Collapse
Affiliation(s)
- Nicholas J Cutrufello
- Bascom Palmer Eye Institute, Department of Ophthalmology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | | | | | |
Collapse
|
23
|
Hu G, Xu C, Staudinger JL. Pregnane X receptor is SUMOylated to repress the inflammatory response. J Pharmacol Exp Ther 2010; 335:342-50. [PMID: 20719936 DOI: 10.1124/jpet.110.171744] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Long-term treatment of patients with the macrolide antibiotic and prototypical activator of pregnane X receptor (PXR) rifampicin (Rif) inhibits the inflammatory response in liver. We show here that activation of the inflammatory response in hepatocytes strongly modulates SUMOylation of ligand-bound PXR. We provide evidence that the SUMOylated PXR contains SUMO3 chains, and feedback represses the immune response in hepatocytes. This information represents the first step in developing novel pharmaceutical strategies to treat inflammatory liver disease and prevent adverse drug reactions in patients experiencing acute or systemic inflammation. These studies also provide a molecular rationale for constructing a novel paradigm that uniquely defines the molecular basis of the interface between PXR-mediated gene activation, drug metabolism, and inflammation.
Collapse
Affiliation(s)
- Gang Hu
- Pharmacology and Toxicology, University of Kansas, 1251 Wescoe Hall Dr., 5038a Malott Hall, Lawrence, KA 66045, USA
| | | | | |
Collapse
|
24
|
Fekih L, Boussoffara L, Jemaa M, Fenniche S, Hassene H, Belhabib D, Megdiche ML. [Tuberculosis in patients with asthma]. Rev Mal Respir 2010; 27:679-84. [PMID: 20863967 DOI: 10.1016/j.rmr.2010.06.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Accepted: 12/03/2009] [Indexed: 11/15/2022]
Abstract
INTRODUCTION The association of asthma and tuberculosis is rare but may raise particular issues around patient management. The aim of this study was to evaluate the clinical, therapeutic and progress of this association. PATIENTS AND METHODS We describe a retrospective study, which included seven asthmatic patients hospitalized for pulmonary tuberculosis during the period between June 2001 and June 2006. RESULTS Five men and two women were included. Mean age was 37 years. Two patients had mild asthma, four had moderate asthma and one had severe and corticosteroid-dependant asthma. Only four patients had controlled asthma when tuberculosis diagnosis was established. Asthma treatment was based on inhaled corticoids and long-acting beta-2-agonists. During antituberculosis treatment two patients developed near fatal asthma. Long-term stable asthma control was achieved over a time course of 3 to 8 years. CONCLUSION The association of asthma and tuberculosis can lead to potential therapeutic difficulties because of pharmacologic interactions between antituberculosis therapies and treatments for asthma treatment. In addition asthma following treatment for tuberculosis appears to be well controlled.
Collapse
Affiliation(s)
- L Fekih
- Service de pneumologie Ibn nafiss, hôpital Abderrahmen Mami, Ariana, Tunisie.
| | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
The immunosuppressive properties of rifampicin have been discussed in the literature for more than 30 years. It is hypothesized that rifampicin acts as a mild immunosuppressive agent in psoriasis rather than an antibacterial one. We report our studies on the therapeutic efficacy of rifampicin in guttate psoriasis. We try to give light on the mechanism of action of rifampicin in psoriasis. Our therapeutic results together with data from the literature revealed rifampicin to be a mild immunosuppressive agent. Its best effectiveness is in the guttate type of the disease.
Collapse
Affiliation(s)
- Nikolai Tsankov
- Department of Dermatology and Venereology, Medical Faculty - Sofia, Bulgaria
| | | |
Collapse
|
26
|
Dubrac S, Elentner A, Ebner S, Horejs-Hoeck J, Schmuth M. Modulation of T lymphocyte function by the pregnane X receptor. THE JOURNAL OF IMMUNOLOGY 2010; 184:2949-57. [PMID: 20173028 DOI: 10.4049/jimmunol.0902151] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The pregnane X receptor (PXR) is a ligand-activated transcription factor regulating genes central to drug and hormone metabolism in the liver. Previous reports indicated that PXR is expressed in PBMC, but the role of PXR in immune cells remains unknown. In this paper, we report increased PXR expression in mouse and human T lymphocytes upon immune activation. Furthermore, pharmacologic activation of PXR inhibits T lymphocyte proliferation and anergizes T lymphocytes by decreasing the expression of CD25 and IFN-gamma and decreasing phosphorylated NF-kappaB and MEK1/2. Although these effects are preceded by an increase of suppressor of cytokine signaling 1, a master switch for IFN-gamma expression, in a PXR-dependent manner, T-bet expression remains unchanged. Conversely, PXR-deficient mice exhibit an exaggerated T lymphocyte proliferation and increased CD25 expression. Furthermore, PXR-deficient lymphocytes produce more IFN-gamma and less of the anti-inflammatory cytokine IL-10. In summary, these results reveal a novel immune-regulatory role of PXR in T lymphocytes and identify suppressor of cytokine signaling 1 as an early signal in PXR-mediated T lymphocyte suppression.
Collapse
Affiliation(s)
- Sandrine Dubrac
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | | | | | | | | |
Collapse
|
27
|
Abassi YA, Xi B, Zhang W, Ye P, Kirstein SL, Gaylord MR, Feinstein SC, Wang X, Xu X. Kinetic cell-based morphological screening: prediction of mechanism of compound action and off-target effects. ACTA ACUST UNITED AC 2009; 16:712-23. [PMID: 19635408 DOI: 10.1016/j.chembiol.2009.05.011] [Citation(s) in RCA: 226] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Revised: 05/29/2009] [Accepted: 05/29/2009] [Indexed: 01/25/2023]
Abstract
We describe a cell-based kinetic profiling approach using impedance readout for monitoring the effect of small molecule compounds. This noninvasive readout allows continuous sampling of cellular responses to biologically active compounds and the ensuing kinetic profile provides information regarding the temporal interaction of compounds with cells. The utility of this approach was tested by screening a library containing FDA approved drugs, experimental compounds, and nature compounds. Compounds with similar activity produced similar impedance-based time-dependent cell response profiles (TCRPs). The compounds were clustered based on TCRP similarity. We identified novel mechanisms for existing drugs, confirmed previously reported calcium modulating activity for COX-2 inhibitor celecoxib, and identified an additional mechanism for the experimental compound monastrol. We also identified and characterized a new antimitotic agent. Our findings indicate that the TCRP approach provides predictive mechanistic information for small molecule compounds.
Collapse
Affiliation(s)
- Yama A Abassi
- ACEA Biosciences, 6779 Mesa Ridge Road, San Diego, CA 92121, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Roles of NF-kappaB activation and peroxisome proliferator-activated receptor gamma inhibition in the effect of rifampin on inducible nitric oxide synthase transcription in human lung epithelial cells. Antimicrob Agents Chemother 2008; 53:1539-45. [PMID: 19114679 DOI: 10.1128/aac.00961-08] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Rifampin (rifampicin), an important antibiotic agent and a major drug used for the treatment of tuberculosis, exerts immunomodulatory effects. Previous studies have found that rifampin increases inducible nitric oxide (NO) synthase (iNOS) expression and NO production. The present study investigated the potential mechanism(s) underlying these actions. The incubation of human lung epithelial A549 cells with a cytokine mix (interleukin-1beta, tumor necrosis factor alpha, and gamma interferon) induced the expression of iNOS mRNA. The addition of rifampin increased the iNOS level by 1.9 +/- 0.3-fold at a dose of 10 microg/ml (P < 0.01) and by 4.0 +/- 0.3-fold at a dose of 50 microg/ml (P < 0.001). Rifampin treatment also affected the transcription factors that regulate iNOS mRNA: there was an increased and prolonged degradation of the inhibitory subunit of NF-kappaB, a corresponding increase in the level of cytokine-induced DNA binding of NF-kappaB (2.1 +/- 0.2-fold), and a decrease in the level of expression of peroxisome proliferator-activated receptor gamma (PPARgamma). Specifically, the level of PPARgamma expression dropped by 15% in response to cytokine stimulation and by an additional 40% when rifampin was added (P < 0.001). Rifampin had no effect on the activation of mitogen-activated protein kinases or the signal transducer and transcription activator (STAT-1). In conclusion, rifampin augments NO production by upregulating iNOS mRNA. It also increases the level of NF-kappaB activation and decreases the level of PPARgamma expression. The increases in the levels of NF-kappaB activation and NO production probably contribute to the therapeutic effects of rifampin. However, given the role of NF-kappaB in upregulating many inflammatory genes and the roles of PPARgamma in downregulating inflammatory genes and in lipid and glucose metabolism, these findings have implications for potential adverse effects of rifampin in patients with chronic inflammatory diseases and glucose or lipid disorders.
Collapse
|
29
|
|
30
|
Yuhas Y, Azoulay-Alfaguter I, Berent E, Ashkenazi S. Rifampin inhibits prostaglandin E2 production and arachidonic acid release in human alveolar epithelial cells. Antimicrob Agents Chemother 2007; 51:4225-30. [PMID: 17908941 PMCID: PMC2168000 DOI: 10.1128/aac.00985-07] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Rifampin, a potent antimicrobial agent, is a major drug in the treatment of tuberculosis. There is evidence that rifampin also serves as an immunomodulator. Based on findings that arachidonic acid and its metabolites are involved in the pathogeneses of Mycobacterium tuberculosis infections, we investigated whether rifampin affects prostaglandin E(2) (PGE(2)) production in human alveolar epithelial cells stimulated with interleukin-1beta. Rifampin caused a dose-dependent inhibition of PGE(2) production. At doses of 100, 50, and 25 microg/ml, it inhibited PGE(2) production by 75%, 59%, and 45%, respectively (P < 0.001). Regarding the mechanism involved, rifampin caused a time- and dose-dependent inhibition of arachidonic acid release from the alveolar cells. At doses of 100, 50, 25, and 10 mug/ml, it significantly inhibited the release of arachidonic acid by 93%, 64%, 58%, and 35%, respectively (P < 0.001). Rifampin did not affect the phosphorylation of cytosolic phospholipase A(2) or the expression of cyclooxygenase-2. The inhibition of PGE(2), and presumably other arachidonic acid products, probably contributes to the efficacy of rifampin in the treatment of tuberculosis and may explain some of its adverse effects.
Collapse
Affiliation(s)
- Yael Yuhas
- Laboratory of Infectious Diseases, Felsenstein Medical Research Center, Beilinson Campus, Petach Tikva 49100, Israel.
| | | | | | | |
Collapse
|
31
|
Abstract
PURPOSE The treatment of cholestatic pruritus in children is reviewed. SUMMARY Cholestasis is characterized by an accumulation of substances that are normally secreted in the bile. Pruritus is a well-known feature of chronic cholestasis in both adults and children and has been reported as the most incapacitating symptom in children with chronic liver disease. Traditional agents, such as antihistamines, are typically ineffective as monotherapy in controlling cholestatic pruritus. As a result, clinicians have looked to other agents, such as rifampin, phenobarbital, ursodiol, opioid antagonists, and bile-binding resins, for attaining better control of pruritic symptoms. Each agent demonstrates different levels of efficacy in pediatric and adult literature. There are no guidelines or algorithms to guide therapy with these agents for children. As a result, an agent should be selected based on the patient's concurrent diseases and current medication regimen. Cholestyramine and ursodiol are both safe and inexpensive, with documented efficacy for cholestatic pruritus in children. Because cholestatic pruritus is likely a result of multiple mechanisms, combination therapy with agents that have differing mechanisms of action might be beneficial and could capitalize on potential synergy between the agents used. Future therapy for cholestatic pruritus may include serotonin antagonists, selective serotonin-reuptake inhibitors, and leukotriene antagonists. CONCLUSION Depending on the underlying disease state resulting in cholestasis, phenobarbital, ursodiol, bile sequestering agents, and opioid antagonists appear to be most effective for treating pruritus related to intrahepatic cholestasis. Alternatively, rifampin appears to be the only agent with reported treatment efficacy for pruritus related to extrahepatic cholestasis.
Collapse
Affiliation(s)
- Jeffrey J Cies
- Department of Pharmacy, Temple University Children's Medical Center, Philadelphia, PA 19140, USA.
| | | |
Collapse
|
32
|
Alisky JM. Doxycycline and rifampin could be useful therapeutic agents for Gaucher disease. Mol Genet Metab 2007; 90:112. [PMID: 16919491 DOI: 10.1016/j.ymgme.2006.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2006] [Accepted: 06/29/2006] [Indexed: 11/27/2022]
|
33
|
Zhou C, Tabb MM, Nelson EL, Grün F, Verma S, Sadatrafiei A, Lin M, Mallick S, Forman BM, Thummel KE, Blumberg B. Mutual repression between steroid and xenobiotic receptor and NF-kappaB signaling pathways links xenobiotic metabolism and inflammation. J Clin Invest 2006; 116:2280-2289. [PMID: 16841097 PMCID: PMC1501109 DOI: 10.1172/jci26283] [Citation(s) in RCA: 309] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Accepted: 05/23/2006] [Indexed: 12/15/2022] Open
Abstract
While it has long been known that inflammation and infection reduce expression of hepatic cytochrome P450 (CYP) genes involved in xenobiotic metabolism and that exposure to xenobiotic chemicals can impair immune function, the molecular mechanisms underlying both of these phenomena have remained largely unknown. Here we show that activation of the nuclear steroid and xenobiotic receptor (SXR) by commonly used drugs in humans inhibits the activity of NF-kappaB, a key regulator of inflammation and the immune response. NF-kappaB target genes are upregulated and small bowel inflammation is significantly increased in mice lacking the SXR ortholog pregnane X receptor (PXR), thereby demonstrating a direct link between SXR and drug-mediated antagonism of NF-kappaB. Interestingly, NF-kappaB activation reciprocally inhibits SXR and its target genes whereas inhibition of NF-kappaB enhances SXR activity. This SXR/PXR-NF-kappaB axis provides a molecular explanation for the suppression of hepatic CYP mRNAs by inflammatory stimuli as well as the immunosuppressant effects of xenobiotics and SXR-responsive drugs. This mechanistic relationship has clinical consequences for individuals undergoing therapeutic exposure to the wide variety of drugs that are also SXR agonists.
Collapse
Affiliation(s)
- Changcheng Zhou
- Department of Developmental and Cell Biology,
Department of Medicine, and Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA.
Department of Gene Regulation and Drug Discovery, City of Hope National Medical Center, Beckman Research Institute, Gonda Diabetes Research Center, Duarte, California, USA.
Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Michelle M. Tabb
- Department of Developmental and Cell Biology,
Department of Medicine, and Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA.
Department of Gene Regulation and Drug Discovery, City of Hope National Medical Center, Beckman Research Institute, Gonda Diabetes Research Center, Duarte, California, USA.
Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Edward L. Nelson
- Department of Developmental and Cell Biology,
Department of Medicine, and Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA.
Department of Gene Regulation and Drug Discovery, City of Hope National Medical Center, Beckman Research Institute, Gonda Diabetes Research Center, Duarte, California, USA.
Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Felix Grün
- Department of Developmental and Cell Biology,
Department of Medicine, and Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA.
Department of Gene Regulation and Drug Discovery, City of Hope National Medical Center, Beckman Research Institute, Gonda Diabetes Research Center, Duarte, California, USA.
Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Suman Verma
- Department of Developmental and Cell Biology,
Department of Medicine, and Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA.
Department of Gene Regulation and Drug Discovery, City of Hope National Medical Center, Beckman Research Institute, Gonda Diabetes Research Center, Duarte, California, USA.
Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Asal Sadatrafiei
- Department of Developmental and Cell Biology,
Department of Medicine, and Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA.
Department of Gene Regulation and Drug Discovery, City of Hope National Medical Center, Beckman Research Institute, Gonda Diabetes Research Center, Duarte, California, USA.
Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Min Lin
- Department of Developmental and Cell Biology,
Department of Medicine, and Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA.
Department of Gene Regulation and Drug Discovery, City of Hope National Medical Center, Beckman Research Institute, Gonda Diabetes Research Center, Duarte, California, USA.
Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Shyamali Mallick
- Department of Developmental and Cell Biology,
Department of Medicine, and Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA.
Department of Gene Regulation and Drug Discovery, City of Hope National Medical Center, Beckman Research Institute, Gonda Diabetes Research Center, Duarte, California, USA.
Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Barry M. Forman
- Department of Developmental and Cell Biology,
Department of Medicine, and Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA.
Department of Gene Regulation and Drug Discovery, City of Hope National Medical Center, Beckman Research Institute, Gonda Diabetes Research Center, Duarte, California, USA.
Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Kenneth E. Thummel
- Department of Developmental and Cell Biology,
Department of Medicine, and Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA.
Department of Gene Regulation and Drug Discovery, City of Hope National Medical Center, Beckman Research Institute, Gonda Diabetes Research Center, Duarte, California, USA.
Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology,
Department of Medicine, and Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA.
Department of Gene Regulation and Drug Discovery, City of Hope National Medical Center, Beckman Research Institute, Gonda Diabetes Research Center, Duarte, California, USA.
Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
34
|
Abstract
The therapeutic and prophylactic use of glucocorticoids is widespread due to their powerful anti-inflammatory, antiproliferative and immunomodulatory activity. However, long-term use of these drugs can result in severe dose-limiting side effects. One of the most critical and debilitating side effects is osteoporosis, which leads to increased risk of fractures. Glucocorticoids damage bone through several different mechanisms. The search for novel glucocorticoids that have reduced side effects in bone and other tissues is being driven by the identification of new mechanisms of action of the glucocorticoid receptor. This may facilitate the detection of new, safer therapies with efficacies equivalent to currently prescribed steroids.
Collapse
Affiliation(s)
- Jeffrey N Miner
- Department of Molecular and Cell Biology, Ligand Pharmaceuticals, San Diego, CA 92121, USA
| | | | | |
Collapse
|
35
|
Yuhas Y, Berent E, Ovadiah H, Azoulay I, Ashkenazi S. Rifampin augments cytokine-induced nitric oxide production in human alveolar epithelial cells. Antimicrob Agents Chemother 2006; 50:396-8. [PMID: 16377722 PMCID: PMC1346828 DOI: 10.1128/aac.50.1.396-398.2006] [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: 01/16/2023] Open
Abstract
Rifampin increased nitric oxide production and inducible nitric oxide synthase expression in alveolar cells stimulated with cytokines. Nitric oxide concentrations after induction with cytokines, cytokines with 10 microg/ml rifampin, and cytokines with 50 microg/ml rifampin were 3.2, 4.5, and 8.8 microM, respectively (P < 0.02 versus cytokines alone). This indicates that rifampin modulates the immune response.
Collapse
Affiliation(s)
- Yael Yuhas
- Laboratory of Infectious Diseases, Felsenstein Medical Research Center, Beilinson Campus, Petah Tikva 49100, Israel.
| | | | | | | | | |
Collapse
|
36
|
Oida Y, Kitaichi K, Nakayama H, Ito Y, Fujimoto Y, Shimazawa M, Nagai H, Hara H. Rifampicin attenuates the MPTP-induced neurotoxicity in mouse brain. Brain Res 2006; 1082:196-204. [PMID: 16515773 DOI: 10.1016/j.brainres.2006.01.116] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2005] [Revised: 01/25/2006] [Accepted: 01/28/2006] [Indexed: 10/24/2022]
Abstract
Rifampicin, an antibacterial drug, is highly effective in the treatment of tuberculosis and leprosy. Recently, it has been reported to have neuroprotective effects in in vitro and in vivo models. This study was designed to elucidate its neuroprotective effects against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity (known as an in vivo mouse model of Parkinson's disease). Mice were injected intraperitoneally (i.p.) with MPTP (10 mg/kg) four times at 1-h intervals, and brains were analyzed 3 or 7 days later. Rifampicin at 20 mg/kg (i.p., twice) had protective effects against MPTP-induced neuronal damage (immunohistochemical changes in tyrosine hydroxylase) in both the substantia nigra and striatum. Rifampicin also protected against the MPTP-induced depletions of dopamine, 3,4-dihydroxyphenylacetic acid (DOPAC), and homovanillic acid (HVA) in the striatum. The maximal concentrations of rifampicin between 30 and 240 min after a single rifampicin injection (20 mg/kg, i.p.) were 2.6 microM (at 30 min) in plasma and 0.77 microM (at 60 min) in striatum. Next, the effects of rifampicin on oxidative stress [lipid peroxidation in mouse brain homogenates and free radical-scavenging activity against diphenyl-p-picrylhydrazyl (DPPH)] were evaluated to clarify the underlying mechanism. At 1 microM or more, rifampicin significantly inhibited both lipid peroxidation in the striatum and free radical production. These findings suggest that in mice, rifampicin can reach brain tissues at concentrations sufficient to attenuate MPTP-induced neurodegeneration in the nigrostriatal dopaminergic neuronal pathway, and that an inhibitory effect against oxidative stress may be partly responsible for its observed neuroprotective effects.
Collapse
Affiliation(s)
- Y Oida
- Department of Biofunctional Molecules, Gifu Pharmaceutical University, 5-6-1 Mitahora-higashi, Gifu 502-8585, Japan
| | | | | | | | | | | | | | | |
Collapse
|
37
|
dos Santos FDJN, Ximenes VF, da Fonseca LM, de Faria Oliveira OMM, Brunetti IL. Horseradish peroxidase-catalyzed oxidation of rifampicin: reaction rate enhancement by co-oxidation with anti-inflammatory drugs. Biol Pharm Bull 2006; 28:1822-6. [PMID: 16204929 DOI: 10.1248/bpb.28.1822] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The tuberculostatic drug rifampicin has been described as a scavenger of reactive species. Additionally, the recent demonstration that oral therapy with a complex of rifampicin and horseradish peroxidase (HRP) was more effective than rifampicin alone, in an animal model of experimental leprosy, suggested the importance of redox reactions involving rifampicin and their relevance to the mechanism of action. Hence, we studied the oxidation of rifampicin catalyzed by HRP, since this enzyme may represent the prototype of peroxidation-mediated reactions. We found that the antibiotic is efficiently oxidized and that rifampicin-quinone is the product, in a reaction dependent on both HRP and hydrogen peroxide. The steady-state kinetic constants Km(app) (101+/-23 micromol/l), Vmax(app) (0.78+/-0.09 micromol/l.s(-1)) and kcat (5.1+/-0.6 s(-1)) were measured (n=4). The reaction rate was increased by the addition of co-substrates such as tetramethylbenzidine, salicylic acid, 5-aminosalicylic acid and paracetamol. This effect was explained by invoking an electron-transfer mechanism by which these drugs acted as mediators of rifampicin oxidation. We suggested that this drug interaction might be important at the inflammatory site.
Collapse
|
38
|
Abstract
Steroidal glucocorticoids are commonly used due to their powerful antiinflammatory activity. However, despite their excellent efficacy, severe side effects frequently limit the use of these drugs. The search for novel glucocorticoids with reduced side effects has been intensified by the discovery of new molecular details regarding the function of the glucocorticoid receptor. These new insights may pave the way for novel, safer therapies that retain the efficacy of currently prescribed steroids.
Collapse
Affiliation(s)
- Jonathan Rosen
- Department of Molecular and Cell Biology, Ligand Pharmaceuticals, 10275 Science Center Drive, San Diego, California 92121, USA
| | | |
Collapse
|
39
|
Affiliation(s)
- Heinz G Floss
- Department of Chemistry, Box 351700, University of Washington, Seattle, Washington 98195-1700, USA
| | | |
Collapse
|
40
|
Abstract
Inflammation represents a complex biologic and biochemical process involving cells of the immune system and a plethora of biologic mediators in response to mechanical, chemical or infectious injuries. When mobilization of effector cells and molecules becomes excessive, the beneficial aspect of this response--to limit damage and promote healing, can be overriden, resulting in host-cell and tissue dysfunction. Based on the hypothesis that chronic infections underly some inflammatory diseases, antibacterial therapy has long been assessed in various inflammatory settings. Recently, the anti-inflammatory activity of some antibacterial agents has also been suspected. Of these duel-action drugs, ansamycins represent an interesting family. Although their therapeutic use is restricted to potentially infectious inflammatory diseases, many experimental data suggest that these drugs also possess direct inhibitory activity on some crucial proinflammatory effectors. To date, the potent antimycobacterial activity of the therapeutically useful ansamycins precludes their widespread use in inflammatory diseases. However, biosynthetic manipulation remains an attractive route for the generation of pharmacologically useful analogs.
Collapse
Affiliation(s)
- Marie-Thérèse Labro
- INSERM, U479, CHU Xavier Bichat, 16 rue Henri Huchard, 75018, Paris, France.
| |
Collapse
|
41
|
|
42
|
Snell NJ. The treatment of tuberculosis: current status and future prospects. Expert Opin Investig Drugs 2005; 7:545-52. [PMID: 15991992 DOI: 10.1517/13543784.7.4.545] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Although a vaccine and effective chemotherapy against tuberculosis (TB) have been available for more than half a century, TB was declared a global emergency in 1993. Current chemotherapeutic regimens are being undermined by lack of resources for proper implementation and control, and the emergence of multi-drug resistant strains of Mycobacterium tuberculosis. Several new chemotherapeutic agents are under development, mainly derived from existing anti-TB drugs or broad-spectrum antibiotics. New experimental agents include immunomodulants and drugs directed against novel cellular targets.
Collapse
Affiliation(s)
- N J Snell
- European Medical Affairs Department, Bayer Pharma, Stoke Court, Stoke Poges, SL2 4LY, UK
| |
Collapse
|
43
|
Böttcher T, Ren H, Goiny M, Gerber J, Lykkesfeldt J, Kuhnt U, Lotz M, Bunkowski S, Werner C, Schau I, Spreer A, Christen S, Nau R. Clindamycin is neuroprotective in experimental Streptococcus pneumoniae meningitis compared with ceftriaxone. J Neurochem 2004; 91:1450-60. [PMID: 15584921 DOI: 10.1111/j.1471-4159.2004.02837.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In animal models of Streptococcus pneumoniae meningitis, rifampin is neuroprotective in comparison to ceftriaxone. So far it is not clear whether this can be generalized for other protein synthesis-inhibiting antimicrobial agents. We examined the effects of the bactericidal protein synthesis-inhibiting clindamycin (n = 12) on the release of proinflammatory bacterial components, the formation of neurotoxic compounds and neuronal injury compared with the standard therapy with ceftriaxone (n = 12) in a rabbit model of pneumococcal meningitis. Analysis of the CSF and histological evaluation were combined with microdialysis from the hippocampal formation and the neocortex. Compared with ceftriaxone, clindamycin reduced the release of lipoteichoic acids from the bacteria (p = 0.004) into the CSF and the CSF leucocyte count (p = 0.011). This led to lower extracellular concentrations of hydroxyl radicals (p = 0.034) and glutamate (p = 0.016) in the hippocampal formation and a subsequent reduction of extracellular glycerol levels (p = 0.018) and neuronal apoptosis in the dentate gyrus (p = 0.008). The present data document beneficial effects of clindamycin compared with ceftriaxone on various parameters linked with the pathophysiology of pneumococcal meningitis and development of neuronal injury. This study suggests neuroprotection to be a group effect of bactericidal protein synthesis-inhibiting antimicrobial agents compared with the standard therapy with beta-lactam antibiotics in meningitis.
Collapse
Affiliation(s)
- Tobias Böttcher
- Department of Neurology, University of Rostock, Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Rifampicin is an antibacterial agent that is widely used in tuberculosis and leprosy therapy. Interestingly, some experimental studies indicate that rifampicin acts as a hydroxyl radical scavenger and a glucocorticoid receptor activator. In this study, the neuroprotective effect of rifampicin was evaluated after transient and permanent focal cerebral ischemia. Anaesthetized male C57BL/6j mice were submitted to permanent or transient thread occlusion of the middle cerebral artery (MCA). Reperfusion in transient ischemia was initiated 30 min later by thread retraction. Rifampicin or vehicle were applied intraperitoneally before permanent or immediately after 30 min of transient ischemia. Later, 24 h after permanent or transient ischemia, animals were re-anesthetized and decapitated. Brain injury was evaluated by triphenyltetrazolium chloride staining (TTC), terminal transferase biotinylated-dUTP nick end labeling (TUNEL) and cresyl violet staining. A 20-mg/kg sample of rifampicin showed a significant neuroprotection after cerebral ischemia. The number of TUNEL-positive cells in the striatum, where disseminated tissue injury was observed, was also reduced by application of rifampicin as compared with vehicle-treated animals. The present report shows that administration of rifampicin efficiently reduces brain injury after permanent and transient focal cerebral ischemia in mice.
Collapse
Affiliation(s)
- Burak Yulug
- Department of Neurology, University of Dokuz Eylül, TR-35340, Izmir, Turkey
| | | | | | | |
Collapse
|
45
|
Mlambo G, Sigola LB. Rifampicin and dexamethasone have similar effects on macrophage phagocytosis of zymosan, but differ in their effects on nitrite and TNF-alpha production. Int Immunopharmacol 2003; 3:513-22. [PMID: 12689656 DOI: 10.1016/s1567-5769(03)00022-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The antibiotic rifampicin is used extensively in the treatment of mycobacterial and other infections. It has previously been suggested that rifampicin binds to and activates the glucocorticoid receptor potentially leading to pharmacological glucocorticoid-like effects such as host immunosuppression (Calleja et al.). This study compares the effects of rifampicin with the synthetic glucocorticoid dexamethasone, on macrophage phagocytosis of zymosan particles and production of nitric oxide (NO) and tumour necrosis factor-alpha (TNF-alpha) by splenocytes or macrophages. Rifampicin and dexamethasone, partially suppressed zymosan phagocytosis by macrophages, respectively, and both effects were ameliorated by the glucocorticoid receptor antagonist RU486. In other experiments, rifampicin had no effects on NO responses; however, dexamethasone inhibited NO in an RU486-sensitive manner. At high doses, rifampicin moderately suppressed TNF-alpha while dexamethasone inhibited it in a dose-dependent manner, which was ameliorated by the presence of RU486. These findings suggest that rifampicin has differential immunomodulatory effects on these innate immune mechanisms.
Collapse
Affiliation(s)
- Godfree Mlambo
- Department of Immunology, Faculty of Medicine, University of Zimbabwe, PO Box A 178 Avondale, Harare, Zimbabwe
| | | |
Collapse
|
46
|
Abstract
Five decades of experimental and clinical experience have changed corticoid therapy thoroughly. Corticoides have two modes of action. The first is a genomic effect through which anti-inflammatory proteins are formed which inhibit pro-inflammatory cytokines. This effect is initiated even by small doses, but is of late onset. The use of high doses initiates non-genomic effects through alterations of the cell membrane; these effects are found early after initiation of treatment. The risk of adverse corticoid effects are extremely rare when modern application forms and therapy regimens are used: Very high doses for a short time in case of acute states of illness, very low doses in long-term therapy of chronic illnesses, and the use of topical substances wherever this is possible. As for the dose regimen, one should start with an initial dose which suffices to treat the acute state, and subsequently reduce the dosage after the first positive results are obtained. In long-term therapy a daily dose of 5 mg prednisolone should not be exceeded; usually even lower doses are sufficient. These very low doses can only be reached by reducing in steps of one half to one milligram over very long periods of time. During long-term therapy osteoporosis prophylaxis is mandatory. Due to these new therapeutic concepts treatment of rheumatoid arthritis with corticoids is experiencing a revival. Low-dose corticoid therapy is of lower risk than nonsteroidal antirheumatic treatment and slows down disease progression, i.e. joint destruction is significantly inhibited. Corticoids have also undergone a new development in the treatment of asthma. Previously used only in acute systemic therapy, they have now been established in basic therapy, i.e. long term therapy using special topic applications.
Collapse
|
47
|
Gollapudi S, Jaidka S, Gupta S. Molecular basis of rifampicin-induced inhibition of anti-CD95-induced apoptosis of peripheral blood T lymphocytes: the role of CD95 ligand and FLIPs. J Clin Immunol 2003; 23:11-22. [PMID: 12645856 DOI: 10.1023/a:1021944113833] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Rifampicin modulates immune response; however, mechanisms by which it exerts these effects are incompletely understood. Recently, rifampicin has been shown to bind to and activate glucocorticoid receptors. Because of the evidence for a role of glucocorticoids in lymphocyte apoptosis, we hypothesized that rifampicin may exert its influence on the immune system by regulating apoptosis. Therefore, we examined the effect of rifampicin on signaling pathway of anti-CD95-induced apoptosis in peripheral blood lymphocytes. Rifampicin, in a concentration-dependent manner, inhibited anti-CD95-induced apoptosis in both CD4+ and CD8+ T cells, which was associated with the inhibition of activation of both caspase-3 and caspase-8. In addition, rifampicin down-regulated the expression of CD95L and Bax. The inhibitory effects of rifampicin on apoptosis and caspase activation as well as its effect on the expression of CD95L and FLIPs were reversed by RU486, an antagonist of glucocorticoid receptor. These data suggest that rifampicin inhibits anti-CD95-mediated apoptosis in lymphocytes by modulating the expression of certain proteins that regulate apoptosis, at least in part, via glucocorticoid receptors.
Collapse
Affiliation(s)
- Sastry Gollapudi
- Laboratory of Cellular Immunology, Division of Basic and Clinical Immunology, University of California, Irvine, California 92697, USA.
| | | | | |
Collapse
|
48
|
Wanchu A, Bhatnagar A, Khullar M, Sud A, Bambery P, Singh S. Antitubercular therapy decreases nitric oxide production in HIV/TB coinfected patients. BMC Infect Dis 2002; 2:15. [PMID: 12147177 PMCID: PMC119853 DOI: 10.1186/1471-2334-2-15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2002] [Accepted: 07/29/2002] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Nitric oxide (NO) production is increased among patients with human immunodeficiency virus (HIV) infection and also those with tuberculosis (TB). In this study we sought to determine if there was increased NO production among patients with HIV/TB coinfection and the effect of four weeks chemotherapy on this level. PATIENTS AND METHODS 19 patients with HIV/TB coinfection were studied. They were treated with standard four drug antitubercular therapy and sampled at baseline and four weeks. 20 patients with HIV infection but no opportunistic infections were disease controls and 20 individuals as healthy controls. Nitrite and citrulline, surrogate markers for NO, were measured it spectrophotometrically. RESULTS Mean age of HIV/TB patients was 28.4+6.8 years and CD4 count was 116+36.6/mm3. Mean nitrite level among HIV/TB coinfected was 207.6+48.8 nmol/ml. This was significantly higher than 99.7+26.5 nmol/ml, the value for HIV infected without opportunistic infections and 46.4+16.2 nmol/ml, the value for healthy controls (p value <0.01). Level of HIV/TB coinfected declined to 144.5+ 34.4 nmol/ml at four weeks of therapy (p value < 0.05). Mean citrulline among HIV/TB coinfected was 1446.8+468.8 nmol/ml. This was significantly higher than 880.8+ 434.8 nmol/ml, the value for HIV infected without opportunistic infections and 486.6+212.5 nmol/ml, the value for healthy controls (p value <0.01). Levels of HIV/TB infected declined to 1116.2+388.6 nmol/ml at four weeks of therapy (p value <0.05). CONCLUSION NO production is elevated among patients with HIV infection, especially so among HIV/TB coinfected, but declines significantly following 4 weeks of antitubercular therapy.
Collapse
Affiliation(s)
- A Wanchu
- Department of Internal Medicine, Post-Graduate Institute of Medical Education and Research, Chandigarh, INDIA
| | - A Bhatnagar
- Department of Experimental Medicine and Biotechnology, Post-Graduate Institute of Medical Education and Research, Chandigarh, INDIA
| | - M Khullar
- Department of Experimental Medicine and Biotechnology, Post-Graduate Institute of Medical Education and Research, Chandigarh, INDIA
| | - A Sud
- Department of Internal Medicine, Post-Graduate Institute of Medical Education and Research, Chandigarh, INDIA
| | - P Bambery
- Department of Internal Medicine, Post-Graduate Institute of Medical Education and Research, Chandigarh, INDIA
| | - S Singh
- Department of Internal Medicine, Post-Graduate Institute of Medical Education and Research, Chandigarh, INDIA
| |
Collapse
|
49
|
Yerramasetti R, Gollapudi S, Gupta S. Rifampicin inhibits CD95-mediated apoptosis of Jurkat T cells via glucocorticoid receptors by modifying the expression of molecules regulating apoptosis. J Clin Immunol 2002; 22:37-47. [PMID: 11958592 DOI: 10.1023/a:1014256603539] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Rifampicin and its analogues are increasingly used in the treatment and prophylaxis of mycobacterial infections. Recently, it has been demonstrated that rifampicin binds to and activates glucocorticoid receptors (GR). Glucocorticoids may inhibit or promote apoptosis in various cell types. Therefore, we investigated the effect of rifampicin on anti-CD95-induced apoptosis in Jurkat T cells. Rifampicin, in a concentration-dependent manner, inhibited anti-CD95-induced apoptosis. Furthermore, rifampicin down-regulated the expression of Bax and CD95L and up-regulated the expression of Bcl-2, Bcl-xL, and Flice-inhibitory protein-L (FLIPL); however, rifampicin had no effect on CD95 or XIAP expression. Rifampicin did not inhibit the binding of anti-CD95 monoclonal antibody to CD95 receptor. A GR-specific antagonist RU480 reversed the inhibition of apoptosis by rifampicin. Furthermore, rifampicin failed to inhibit anti-CD95-induced apoptosis in a dominant negative IKBalpha (IKBaM) Jurkat T cells. Taken together, these findings suggest that rifampicin inhibits anti-CD95-induced apoptosis in Jurkat T cells by modulating the expression of various molecules regulating apoptosis and its effect appears to be mediated via GR and at least in part through NF-kappaB signaling pathway.
Collapse
Affiliation(s)
- Rama Yerramasetti
- Division of Basic and Clinical Immunology University of California, Irvine 92697, USA
| | | | | |
Collapse
|
50
|
Tiemessen CT, Shalekoff S, Meddows-Taylor S, Martin DJ. Antituberculosis treatment: increasing evidence for drug effects on innate cellular immunity. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2001; 8:686-9. [PMID: 11427411 PMCID: PMC96127 DOI: 10.1128/cdli.8.4.686-689.2001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- C T Tiemessen
- AIDS Virus Research Unit, National Institute for Virology, Johannesburg, South Africa.
| | | | | | | |
Collapse
|