1
|
El-Shoura EAM, Mohamed AAN, Atwa AM, Salem EA, Sharkawi SMZ, Mostafa Selim H, Ibrahim Elberri A, Gawesh ES, Ahmed YH, Abd El-Ghafar OAM. Combined diosmin and bisoprolol attenuate cobalt chloride-induced cardiotoxicity and endothelial dysfunction through modulating miR-143-3P/MAPK/MCP-1, ERK5/CXCR4, Orai-1/STIM-1 signaling pathways. Int Immunopharmacol 2024; 140:112777. [PMID: 39088923 DOI: 10.1016/j.intimp.2024.112777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/05/2024] [Accepted: 07/23/2024] [Indexed: 08/03/2024]
Abstract
Even while accelerated cardiomyocyte apoptosis is one of the primary causes of cardiac damage, the underlying mechanism is still mostly unknown. In addition to examining potential protective effects of bisoprolol and diosmin against CoCl2-induced cardiac injury, the goal of this study was to identify potential mechanisms regulating the hypoxic cardiac damage caused by cobalt chloride (CoCl2). For a period of 21 days except Cocl2 14 days from the first day of the experiment, rats were split into the following groups: Normal control group, rats received vehicle only (2 ml/kg/day, p.o.), (Cocl2, 150 mg/kg/day, p.o.), bisoprolol (25 mg/kg/day, p.o.); diosmin (100 mg/kg/day, p.o.) and bisoprolol + diosmin + Cocl2 groups. At the end of the experimental period, serum was taken for estimation of cardiac function, lipid profile, and pro/anti-inflammatory cytokines. Moreover, tissue samples were collected for evaluation of oxidative stress, endothelial dysfunction, α-SMA, PKC-α, MiR-143-3P, MAPK, ERK5, MCP-1, CXCR4, Orai-1, and STIM-1. Diosmin and bisoprolol, either alone or in combination, enhance heart function by reducing abnormalities in the electrocardiogram and the hypotension brought on by CoCl2. Additionally, they significantly ameliorate endothelial dysfunction by downregulating the cardiac expressions of α-SMA, PKC-α, MiR-143-3P, MAPK, ERK5, MCP-1, CXCR4, Orai-1, and STIM-1. Bisoprolol and diosmin produced modulatory activity against inflammatory state, redox balance, and atherogenic index concurrently. Together, diosmin and bisoprolol, either alone or in combination, significantly reduced all the cardiac alterations brought on by CoCl2. The capacity to obstruct hypoxia-induced α-SMA, PKC-α, MiR-143-3P/MAPK/MCP-1, MiR-143-3P/ERK5/CXCR4, Orai-1/STIM-1 signaling activation, as well as their anti-inflammatory, antioxidant, and anti-apoptotic properties, may be responsible for these cardio-protective results.
Collapse
Affiliation(s)
- Ehab A M El-Shoura
- Clinical Pharmacy Department, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | | | - Ahmed M Atwa
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Esraa A Salem
- Department of Clinical Physiology, Faculty of Medicine, Menoufia University, Shebeen ElKom, 32511, Egypt
| | - Souty M Z Sharkawi
- Pharmacology and Toxicology Department, Beni Suef University, Beni Suef, Egypt
| | | | - Aya Ibrahim Elberri
- Genetic Engineering and Molecular Biology Division, Department of Zoology, Faculty of Science, Menoufia University, Shebeen Elkom 32511, Egypt
| | - El-Sayed Gawesh
- Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Yasmine H Ahmed
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | | |
Collapse
|
2
|
Sales ALCC, Primo MGS, Mello Neto RS, Mendes AVS, Furtado MM, Rocha JÉL, Almeida JOCS, França JVS, Alencar SR, Brito AKS, Lopes LO, Rizzo MS, Lustosa AKMF, Nunes PHM, Lucarini M, Durazzo A, Arcanjo DDR, Martins MDCC. A Preliminary Study on Hepatoprotective, Hypolipidemic and Aortic Morphometric Effects of Omega-3-Rich Fish Oil in Hypercholesterolemic Mice. Pharmaceuticals (Basel) 2024; 17:72. [PMID: 38256905 PMCID: PMC10819677 DOI: 10.3390/ph17010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
This study aims to evaluate the hepatoprotective, hypolipidemic and aortic morphometric effects of fish oil rich in omega-3 in hypercholesterolemic BALB/c mice. This is an experimental model that included 16 male BALB/c mice (Mus musculus) divided into three groups (G1 (standard commercial chow and 0.9% saline solution), G2 (hypercholesterolemic diet and 0.9% saline solution) and G3 (hypercholesterolemic diet and fish oil)) for 8 weeks. There was no significant difference in the treatment with omega-3-rich fish oil in the lipid profile (p > 0.05). In the histological analysis, group G2 detected the presence of hepatitis and liver tissue necrosis, but this was not observed in group G3. As for the morphometry in the light area of the vessel, the G1 group had a higher score (2.62 ± 0.36 mm2) when compared to G2 (2.10 ± 0.16 mm2) and G3 (2.26 ± 0.25 mm2) (p < 0.05). The vessel wall thickness did not differ between the groups (p > 0.05). It is concluded that supplementation with fish oil rich in omega-3 carried out in this study may have a protective effect on liver tissue, but it has not yet improved the lipid and morphometric profile. Despite this research being preliminary, it is a relevant study with future prospects for improving the doses of EPA and DHA in order to better elucidate the benefits of fish oil in models of dyslipidemia.
Collapse
Affiliation(s)
- Ana Lina C. C. Sales
- Department of Biophysics and Physiology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil; (A.L.C.C.S.); (M.G.S.P.); (R.S.M.N.); (A.V.S.M.); (M.M.F.); (J.É.L.R.); (J.O.C.S.A.); (J.V.S.F.); (S.R.A.); (A.K.S.B.); (P.H.M.N.); (D.D.R.A.)
- University Hospital, Federal University of Piauí, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil;
| | - Maísa G. S. Primo
- Department of Biophysics and Physiology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil; (A.L.C.C.S.); (M.G.S.P.); (R.S.M.N.); (A.V.S.M.); (M.M.F.); (J.É.L.R.); (J.O.C.S.A.); (J.V.S.F.); (S.R.A.); (A.K.S.B.); (P.H.M.N.); (D.D.R.A.)
| | - Renato S. Mello Neto
- Department of Biophysics and Physiology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil; (A.L.C.C.S.); (M.G.S.P.); (R.S.M.N.); (A.V.S.M.); (M.M.F.); (J.É.L.R.); (J.O.C.S.A.); (J.V.S.F.); (S.R.A.); (A.K.S.B.); (P.H.M.N.); (D.D.R.A.)
| | - Ana Victória S. Mendes
- Department of Biophysics and Physiology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil; (A.L.C.C.S.); (M.G.S.P.); (R.S.M.N.); (A.V.S.M.); (M.M.F.); (J.É.L.R.); (J.O.C.S.A.); (J.V.S.F.); (S.R.A.); (A.K.S.B.); (P.H.M.N.); (D.D.R.A.)
| | - Mariely M. Furtado
- Department of Biophysics and Physiology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil; (A.L.C.C.S.); (M.G.S.P.); (R.S.M.N.); (A.V.S.M.); (M.M.F.); (J.É.L.R.); (J.O.C.S.A.); (J.V.S.F.); (S.R.A.); (A.K.S.B.); (P.H.M.N.); (D.D.R.A.)
| | - Joana Érica L. Rocha
- Department of Biophysics and Physiology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil; (A.L.C.C.S.); (M.G.S.P.); (R.S.M.N.); (A.V.S.M.); (M.M.F.); (J.É.L.R.); (J.O.C.S.A.); (J.V.S.F.); (S.R.A.); (A.K.S.B.); (P.H.M.N.); (D.D.R.A.)
| | - José Otávio C. S. Almeida
- Department of Biophysics and Physiology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil; (A.L.C.C.S.); (M.G.S.P.); (R.S.M.N.); (A.V.S.M.); (M.M.F.); (J.É.L.R.); (J.O.C.S.A.); (J.V.S.F.); (S.R.A.); (A.K.S.B.); (P.H.M.N.); (D.D.R.A.)
| | - José Vinícius S. França
- Department of Biophysics and Physiology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil; (A.L.C.C.S.); (M.G.S.P.); (R.S.M.N.); (A.V.S.M.); (M.M.F.); (J.É.L.R.); (J.O.C.S.A.); (J.V.S.F.); (S.R.A.); (A.K.S.B.); (P.H.M.N.); (D.D.R.A.)
| | - Salmon R. Alencar
- Department of Biophysics and Physiology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil; (A.L.C.C.S.); (M.G.S.P.); (R.S.M.N.); (A.V.S.M.); (M.M.F.); (J.É.L.R.); (J.O.C.S.A.); (J.V.S.F.); (S.R.A.); (A.K.S.B.); (P.H.M.N.); (D.D.R.A.)
| | - Ana Karolinne S. Brito
- Department of Biophysics and Physiology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil; (A.L.C.C.S.); (M.G.S.P.); (R.S.M.N.); (A.V.S.M.); (M.M.F.); (J.É.L.R.); (J.O.C.S.A.); (J.V.S.F.); (S.R.A.); (A.K.S.B.); (P.H.M.N.); (D.D.R.A.)
| | - Luana O. Lopes
- University Hospital, Federal University of Piauí, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil;
| | - Márcia S. Rizzo
- Department of Morphology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil;
| | - Ana Karina M. F. Lustosa
- Galeno Farmácia de Manipulação, Virgínia Regina Fortes Castelo Branco e Cia. Ltda., Teresina 64001-260, PI, Brazil;
| | - Paulo Humberto M. Nunes
- Department of Biophysics and Physiology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil; (A.L.C.C.S.); (M.G.S.P.); (R.S.M.N.); (A.V.S.M.); (M.M.F.); (J.É.L.R.); (J.O.C.S.A.); (J.V.S.F.); (S.R.A.); (A.K.S.B.); (P.H.M.N.); (D.D.R.A.)
| | - Massimo Lucarini
- CREA-Research Centre for Food and Nutrition, Via Ardeatina 546, 00178 Rome, Italy;
| | - Alessandra Durazzo
- CREA-Research Centre for Food and Nutrition, Via Ardeatina 546, 00178 Rome, Italy;
| | - Daniel Dias Rufino Arcanjo
- Department of Biophysics and Physiology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil; (A.L.C.C.S.); (M.G.S.P.); (R.S.M.N.); (A.V.S.M.); (M.M.F.); (J.É.L.R.); (J.O.C.S.A.); (J.V.S.F.); (S.R.A.); (A.K.S.B.); (P.H.M.N.); (D.D.R.A.)
| | - Maria do Carmo C. Martins
- Department of Biophysics and Physiology, Federal University of Piaui, Campus Ministro Petrônio Portella, Ininga, Teresina 64049-550, PI, Brazil; (A.L.C.C.S.); (M.G.S.P.); (R.S.M.N.); (A.V.S.M.); (M.M.F.); (J.É.L.R.); (J.O.C.S.A.); (J.V.S.F.); (S.R.A.); (A.K.S.B.); (P.H.M.N.); (D.D.R.A.)
| |
Collapse
|
3
|
El-Shoura EAM, Salem MA, Ahmed YH, Ahmed LK, Zaafar D. Combined β-sitosterol and trimetazidine mitigate potassium dichromate-induced cardiotoxicity in rats through the interplay between NF-κB/AMPK/mTOR/TLR4 and HO-1/NADPH signaling pathways. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:67771-67787. [PMID: 37115449 PMCID: PMC10203021 DOI: 10.1007/s11356-023-27021-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/10/2023] [Indexed: 05/25/2023]
Abstract
Hexavalent chromium salt, like potassium dichromate (PD), is chromium's most precarious valence state in industrial wastes. Recently, there has been increasing interest in β-sitosterol (BSS), a bioactive phytosterol, as a dietary supplement. BSS is recommended in treating cardiovascular disorders due to its antioxidant effect. Trimetazidine (TMZ) was used traditionally for cardioprotection. Through the administration of BSS and TMZ, the cardiotoxic effects of PD were to be countered in this study, in addition to examining the precise mechanism of PD-induced cardiotoxicity. Thirty male albino rats were divided into five groups; the control group: administered normal saline daily (3 mL/kg); the PD group: administered normal saline daily (3 mL/kg); BSS group: administered BSS daily (20 mg/kg); TMZ group: administered TMZ daily (15 mg/kg); and the BSS + TMZ group: administered both BSS (20 mg/kg) and TMZ (15 mg/kg) daily. All experimental groups, except the control, received on the 19th day a single dose of PD (30 mg/kg/day, S.C.). Normal saline, BSS, and TMZ were received daily for 21 consecutive days p.o. The exposure to PD promoted different oxidative stresses, pro-inflammatory, and cardiotoxicity biomarkers. BSS or TMZ succeeded solely in reducing these deleterious effects; however, their combination notably returned measured biomarkers close to normal values. The histopathological investigations have supported the biochemical findings. The combination of BSS and TMZ protects against PD cardiotoxicity in rats by reducing oxidative stress and apoptotic and inflammatory biomarkers. It may be promising for alleviating and protecting against PD-induced cardiotoxicity in people at an early stage; however, these findings need further clinical studies to be confirmed. HIGHLIGHTS: • Potassium dichromate induces cardiotoxicity in rats through the upregulation of oxidative stress, proinflammatory, and apoptotic pathways biomarkers. • β-Sitosterol possesses a possible cardioprotective effect by modulating several signaling pathways. • Trimetazidine, the antianginal agent, has a potential cardioprotective impact on PD-intoxicated rat model. • The combination of β-Sitosterol and trimetazidine was the best in modulating different pathways involved in PD cardiotoxicity in rats via the interplay between NF-κB/AMPK/mTOR/TLR4 and HO-1/NADPH signaling pathways.
Collapse
Affiliation(s)
- Ehab A. M. El-Shoura
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch Assiut, 71524 Egypt
| | - Maha A. Salem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology, and Information, Cairo, Egypt
| | - Yasmine H. Ahmed
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Lamiaa Khalaf Ahmed
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 71524 Egypt
| | - Dalia Zaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology, and Information, Cairo, Egypt
| |
Collapse
|
4
|
Gibson BG, Cox TE, Marchbank KJ. Contribution of animal models to the mechanistic understanding of Alternative Pathway and Amplification Loop (AP/AL)-driven Complement-mediated Diseases. Immunol Rev 2023; 313:194-216. [PMID: 36203396 PMCID: PMC10092198 DOI: 10.1111/imr.13141] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
This review aimed to capture the key findings that animal models have provided around the role of the alternative pathway and amplification loop (AP/AL) in disease. Animal models, particularly mouse models, have been incredibly useful to define the role of complement and the alternative pathway in health and disease; for instance, the use of cobra venom factor and depletion of C3 provided the initial insight that complement was essential to generate an appropriate adaptive immune response. The development of knockout mice have further underlined the importance of the AP/AL in disease, with the FH knockout mouse paving the way for the first anti-complement drugs. The impact from the development of FB, properdin, and C3 knockout mice closely follows this in terms of mechanistic understanding in disease. Indeed, our current understanding that complement plays a role in most conditions at one level or another is rooted in many of these in vivo studies. That C3, in particular, has roles beyond the obvious in innate and adaptive immunity, normal physiology, and cellular functions, with or without other recognized AP components, we would argue, only extends the reach of this arm of the complement system. Humanized mouse models also continue to play their part. Here, we argue that the animal models developed over the last few decades have truly helped define the role of the AP/AL in disease.
Collapse
Affiliation(s)
- Beth G. Gibson
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| | - Thomas E. Cox
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| | - Kevin J. Marchbank
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| |
Collapse
|
5
|
Janneh AH, Kassir MF, Atilgan FC, Lee HG, Sheridan M, Oleinik N, Szulc Z, Voelkel-Johnson C, Nguyen H, Li H, Peterson YK, Marangoni E, Saatci O, Sahin O, Lilly M, Atkinson C, Tomlinson S, Mehrotra S, Ogretmen B. Crosstalk between pro-survival sphingolipid metabolism and complement signaling induces inflammasome-mediated tumor metastasis. Cell Rep 2022; 41:111742. [PMID: 36476873 PMCID: PMC9791981 DOI: 10.1016/j.celrep.2022.111742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 08/15/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022] Open
Abstract
Crosstalk between metabolic and signaling events that induce tumor metastasis remains elusive. Here, we determine how oncogenic sphingosine 1-phosphate (S1P) metabolism induces intracellular C3 complement activation to enhance migration/metastasis. We demonstrate that increased S1P metabolism activates C3 complement processing through S1P receptor 1 (S1PR1). S1P/S1PR1-activated intracellular C3b-α'2 is associated with PPIL1 through glutamic acid 156 (E156) and aspartic acid 111 (D111) residues, resulting in NLRP3/inflammasome induction. Inactivation mutations of S1PR1 to prevent S1P signaling or mutations of C3b-α'2 to prevent its association with PPIL1 attenuate inflammasome activation and reduce lung colonization/metastasis in mice. Also, activation of the S1PR1/C3/PPIL1/NLRP3 axis is highly associated with human metastatic melanoma tissues and patient-derived xenografts. Moreover, targeting S1PR1/C3/PPIL1/NLRP3 signaling using molecular, genetic, and pharmacologic tools prevents lung colonization/metastasis of various murine cancer cell lines using WT and C3a-receptor1 knockout (C3aR1-/-) mice. These data provide strategies for treating high-grade/metastatic tumors by targeting the S1PR1/C3/inflammasome axis.
Collapse
Affiliation(s)
- Alhaji H Janneh
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Mohamed Faisal Kassir
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - F Cansu Atilgan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Han Gyul Lee
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Megan Sheridan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Natalia Oleinik
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Zdzislaw Szulc
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Christina Voelkel-Johnson
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Department of Microbiology and Immunology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Hung Nguyen
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Department of Microbiology and Immunology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Hong Li
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Department of Public Health, College of Medicine, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Yuri K Peterson
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | | | - Ozge Saatci
- Department of Drug Discovery and Biomedical Sciences, School of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Ozgur Sahin
- Department of Drug Discovery and Biomedical Sciences, School of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Michael Lilly
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Carl Atkinson
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Department of Microbiology and Immunology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Stephen Tomlinson
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Department of Microbiology and Immunology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Shikhar Mehrotra
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Department of Microbiology and Immunology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA.
| |
Collapse
|
6
|
Immune response associated with ischemia and reperfusion injury during organ transplantation. Inflamm Res 2022; 71:1463-1476. [PMID: 36282292 PMCID: PMC9653341 DOI: 10.1007/s00011-022-01651-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 12/03/2022] Open
Abstract
Background Ischemia and reperfusion injury (IRI) is an ineluctable immune-related pathophysiological process during organ transplantation, which not only causes a shortage of donor organs, but also has long-term and short-term negative consequences on patients. Severe IRI-induced cell death leads to the release of endogenous substances, which bind specifically to receptors on immune cells to initiate an immune response. Although innate and adaptive immunity have been discovered to play essential roles in IRI in the context of organ transplantation, the pathway and precise involvement of the immune response at various stages has not yet to be elucidated. Methods We combined “IRI” and “organ transplantation” with keywords, respectively such as immune cells, danger signal molecules, macrophages, neutrophils, natural killer cells, complement cascade, T cells or B cells in PubMed and the Web of Science to search for relevant literatures. Conclusion Comprehension of the immune mechanisms involved in organ transplantation is promising for the treatment of IRI, this review summarizes the similarities and differences in both innate and adaptive immunity and advancements in the immune response associated with IRI during diverse organ transplantation.
Collapse
|
7
|
Milusev A, Rieben R, Sorvillo N. The Endothelial Glycocalyx: A Possible Therapeutic Target in Cardiovascular Disorders. Front Cardiovasc Med 2022; 9:897087. [PMID: 35647072 PMCID: PMC9136230 DOI: 10.3389/fcvm.2022.897087] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/21/2022] [Indexed: 12/15/2022] Open
Abstract
The physiological, anti-inflammatory, and anti-coagulant properties of endothelial cells (ECs) rely on a complex carbohydrate-rich layer covering the luminal surface of ECs, called the glycocalyx. In a range of cardiovascular disorders, glycocalyx shedding causes endothelial dysfunction and inflammation, underscoring the importance of glycocalyx preservation to avoid disease initiation and progression. In this review we discuss the physiological functions of the glycocalyx with particular focus on how loss of endothelial glycocalyx integrity is linked to cardiovascular risk factors, like hypertension, aging, diabetes and obesity, and contributes to the development of thrombo-inflammatory conditions. Finally, we consider the role of glycocalyx components in regulating inflammatory responses and discuss possible therapeutic interventions aiming at preserving or restoring the endothelial glycocalyx and therefore protecting against cardiovascular disease.
Collapse
Affiliation(s)
- Anastasia Milusev
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Robert Rieben
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Nicoletta Sorvillo
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- *Correspondence: Nicoletta Sorvillo
| |
Collapse
|
8
|
Magalhães LSSM, Andrade DB, Bezerra RDS, Morais AIS, Oliveira FC, Rizzo MS, Silva-Filho EC, Lobo AO. Nanocomposite Hydrogel Produced from PEGDA and Laponite for Bone Regeneration. J Funct Biomater 2022; 13:53. [PMID: 35645261 PMCID: PMC9149996 DOI: 10.3390/jfb13020053] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 01/03/2023] Open
Abstract
Herein, a nanocomposite hydrogel was produced using laponite and polyethylene-glycol diacrylate (PEGDA), with or without Irgacure (IG), for application in bone tissue regeneration. The nanocomposites were characterized by X-ray diffraction (XRD), Fourier-Transform infrared spectroscopy (FTIR), and thermal analysis (TG/DTG). The XRD results showed that the crystallographic structure of laponite was preserved in the nanocomposite hydrogels after the incorporation of PEGDA and IG. The FTIR results indicated that PEGDA polymer chains were entangled on laponite in hydrogels. The TG/DTG found that the presence of laponite (Lap) improved the thermal stability of nanocomposite hydrogel. The toxicity tests by Artemia salina indicated that the nanocomposite hydrogels were not toxic, because the amount of live nauplii was 80.0%. In addition, in vivo tests demonstrated that the hydrogels had the ability to regenerate bone in a bone defect model of the tibiae of osteopenic rats. For the nanocomposite hydrogel (PEGDA + Lap nanocomposites + UV light), the formation of intramembranous bone in the soft callus was more intense in 66.7% of the animals. Thus, the results presented in this study evidence that nanocomposite hydrogels obtained from laponite and PEGDA have the potential for use in bone regeneration.
Collapse
Affiliation(s)
- Leila S. S. M. Magalhães
- LIMAV—Interdisciplinary Advanced Materials Laboratory, PPGCM—Materials Science and Engineering Graduate Program, UFPI—Federal University of Piaui, Teresina 64049-550, Brazil; (L.S.S.M.M.); (D.B.A.); (A.I.S.M.); (M.S.R.); (E.C.S.-F.)
| | - Danielle B. Andrade
- LIMAV—Interdisciplinary Advanced Materials Laboratory, PPGCM—Materials Science and Engineering Graduate Program, UFPI—Federal University of Piaui, Teresina 64049-550, Brazil; (L.S.S.M.M.); (D.B.A.); (A.I.S.M.); (M.S.R.); (E.C.S.-F.)
- Federal Institute of Education, Science and Technology of Piauí, Teresina-Central Campus, IFPI, Teresina 64000-040, Brazil;
| | - Roosevelt D. S. Bezerra
- Federal Institute of Education, Science and Technology of Piauí, Teresina-Central Campus, IFPI, Teresina 64000-040, Brazil;
| | - Alan I. S. Morais
- LIMAV—Interdisciplinary Advanced Materials Laboratory, PPGCM—Materials Science and Engineering Graduate Program, UFPI—Federal University of Piaui, Teresina 64049-550, Brazil; (L.S.S.M.M.); (D.B.A.); (A.I.S.M.); (M.S.R.); (E.C.S.-F.)
| | | | - Márcia S. Rizzo
- LIMAV—Interdisciplinary Advanced Materials Laboratory, PPGCM—Materials Science and Engineering Graduate Program, UFPI—Federal University of Piaui, Teresina 64049-550, Brazil; (L.S.S.M.M.); (D.B.A.); (A.I.S.M.); (M.S.R.); (E.C.S.-F.)
| | - Edson C. Silva-Filho
- LIMAV—Interdisciplinary Advanced Materials Laboratory, PPGCM—Materials Science and Engineering Graduate Program, UFPI—Federal University of Piaui, Teresina 64049-550, Brazil; (L.S.S.M.M.); (D.B.A.); (A.I.S.M.); (M.S.R.); (E.C.S.-F.)
| | - Anderson O. Lobo
- LIMAV—Interdisciplinary Advanced Materials Laboratory, PPGCM—Materials Science and Engineering Graduate Program, UFPI—Federal University of Piaui, Teresina 64049-550, Brazil; (L.S.S.M.M.); (D.B.A.); (A.I.S.M.); (M.S.R.); (E.C.S.-F.)
| |
Collapse
|
9
|
Hamdan DI, Hafez SS, Hassan WHB, Morsi MM, Khalil HMA, Ahmed YH, Ahmed-Farid OA, El-Shiekh RA. Chemical profiles with cardioprotective and anti-depressive effects of Morus macroura Miq. leaves and stem branches dichloromethane fractions on isoprenaline induced post-MI depression. RSC Adv 2022; 12:3476-3493. [PMID: 35425386 PMCID: PMC8979319 DOI: 10.1039/d1ra08320a] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/04/2022] [Indexed: 12/27/2022] Open
Abstract
This study was conducted to explore the potential cardioprotective and anti-depressive effects of dichloromethane (DCM) fractions of Morus macroura leaves (L) and stem branches (S) on post-myocardial infarction (MI) depression induced by isoprenaline (ISO) in rats in relation to their metabolites. The study was propped with a UPLC-ESI-MS/MS profiling and chromatographic isolation of the secondary metabolites. Column chromatography revealed the isolation of lupeol palmitate (6) that was isolated for the first time from nature with eight known compounds. In addition, more than forty metabolites belonging, mainly to flavonoids, and anthocyanins groups were identified. The rats were injected with ISO (85 mg kg−1, s.c) in the first two days, followed by the administration of M. macroura DCM-L and DCM-S fractions (200 mg kg−1 p.o) for 19 days. Compared with the ISO exposed rats, the treated rats displayed a reduction in cardiac biomarkers (LDH and CKMB), anxiety, and depressive-like behaviour associated with an increase in the brain defense system (SOD and GSH), neuronal cell energy, GABA, serotonin, and dopamine, confirmed by histopathological investigations. In conclusion, DCM-L and DCM-S fractions' cardioprotective and anti-depressive activities are attributed to their metabolite profile. Therefore, they could serve as a potential agent in amending post-MI depression. This study was conducted to explore the potential cardioprotective and anti-depressive effects of dichloromethane fractions of Morus macroura leaves and stem branches on post-myocardial infarction depression induced by isoprenaline in rats in relation to their metabolites.![]()
Collapse
Affiliation(s)
- Dalia I Hamdan
- Department of Pharmacognosy and Natural Products, Faculty of Pharmacy, Menoufia University Shibin Elkom 32511 Egypt
| | - Samia S Hafez
- Pharmacognosy Department, Faculty of Pharmacy, Zagazig University Zagazig 44519 Egypt
| | - Wafaa H B Hassan
- Pharmacognosy Department, Faculty of Pharmacy, Zagazig University Zagazig 44519 Egypt
| | - Mai M Morsi
- Pharmacognosy Department, Faculty of Pharmacy, Zagazig University Zagazig 44519 Egypt
| | - Heba M A Khalil
- Department of Veterinary Hygiene and Management, Faculty of Veterinary Medicine, Cairo University Giza 12211 Egypt +201013666331
| | - Yasmine H Ahmed
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University Giza 12211 Egypt
| | - Omar A Ahmed-Farid
- Department of Physiology, National Organization for Drug Control and Research Giza Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University Kasr El Aini St. Cairo 11562 Egypt +201064763764
| |
Collapse
|
10
|
Zheng C, Sleiman MM, Yang X, He S, Atkinson C, Tomlinson S. Increasing the efficacy and safety of a human complement inhibitor for treating post-transplant cardiac ischemia reperfusion injury by targeting to a graft-specific neoepitope. J Heart Lung Transplant 2021; 40:1112-1121. [PMID: 34334299 PMCID: PMC10587835 DOI: 10.1016/j.healun.2021.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 06/17/2021] [Accepted: 07/03/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Post-transplant ischemia reperfusion injury (IRI) is a recognized risk factor for subsequent organ dysfunction, alloresponsiveness, and rejection. The complement system is known to play a role in IRI and represents a therapeutic target. Complement is activated in transplanted grafts when circulating IgM antibodies bind to exposed ischemia-induced neoepitopes upon reperfusion, and we investigated the targeting of a human complement inhibitor, CR1, to a post-transplant ischemia-induced neoepitope. METHODS A fragment of human CR1 was linked to a single chain antibody construct (C2 scFv) recognizing an injury-specific neoepitope to yield C2-CR1. This construct, along with a soluble untargeted counterpart, was characterized in a cardiac allograft transplantation model of IRI in terms of efficacy and safety. RESULTS CR1 was similarly effective against mouse and human complement. C2-CR1 provided effective protection against cardiac IRI at a lower dose than untargeted CR1. The increased efficacy of C2-CR1 relative to CR1 correlated with decreased C3 deposition, and C2-CR1, but not CR1, targeted to cardiac allografts. At a dose necessary to reduce IRI, C2-CR1 had minimal impact on serum complement activity, in contrast to CR1 which resulted in a high level of systemic inhibition. The circulatory half-life of CR1 was markedly longer than that of C2-CR1, and whereas a minimum therapeutic dose of CR1 severely impaired host susceptibility to infection, C2-CR1 had no impact. CONCLUSION We show the translational potential of a human complement inhibitor targeted to a universal ischemia-induced graft-specific epitope, and demonstrate advantages compared to an untargeted counterpart in terms of efficacy and safety.
Collapse
Affiliation(s)
- Chaowen Zheng
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Division of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Mohamad Mahdi Sleiman
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Xiaofeng Yang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Songqing He
- Division of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- The Lee Patterson Allen Transplant Immunobiology Laboratory, Department of Transplant Surgery, Medical University of South Carolina, Department of Surgery, Charleston, SC, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
- Department of Pulmonary Medicine, University of Florida, Gainesville, FL, USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| |
Collapse
|
11
|
Patel PM, Connolly MR, Coe TM, Calhoun A, Pollok F, Markmann JF, Burdorf L, Azimzadeh A, Madsen JC, Pierson RN. Minimizing Ischemia Reperfusion Injury in Xenotransplantation. Front Immunol 2021; 12:681504. [PMID: 34566955 PMCID: PMC8458821 DOI: 10.3389/fimmu.2021.681504] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/12/2021] [Indexed: 12/21/2022] Open
Abstract
The recent dramatic advances in preventing "initial xenograft dysfunction" in pig-to-non-human primate heart transplantation achieved by minimizing ischemia suggests that ischemia reperfusion injury (IRI) plays an important role in cardiac xenotransplantation. Here we review the molecular, cellular, and immune mechanisms that characterize IRI and associated "primary graft dysfunction" in allotransplantation and consider how they correspond with "xeno-associated" injury mechanisms. Based on this analysis, we describe potential genetic modifications as well as novel technical strategies that may minimize IRI for heart and other organ xenografts and which could facilitate safe and effective clinical xenotransplantation.
Collapse
Affiliation(s)
- Parth M. Patel
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Margaret R. Connolly
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Taylor M. Coe
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Anthony Calhoun
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Franziska Pollok
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Anesthesiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - James F. Markmann
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Transplantation, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Lars Burdorf
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Agnes Azimzadeh
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Joren C. Madsen
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Richard N. Pierson
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
12
|
Lei B, Sleiman MM, Cheng Q, Tu Z, Zhu P, Goddard M, Martins PN, Langerude L, Nadig S, Tomlinson S, Atkinson C. In Situ Pre-Treatment of Vascularized Composite Allografts With a Targeted Complement Inhibitor Protects Against Brain Death and Ischemia Reperfusion Induced Injuries. Front Immunol 2021; 12:630581. [PMID: 34394069 PMCID: PMC8358649 DOI: 10.3389/fimmu.2021.630581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
Introduction Donor brain death (BD) is an unavoidable component of vascularized composite allograft (VCA) transplantation and a key contributor to ischemia-reperfusion injury (IRI). Complement is activated and deposited within solid organ grafts as a consequence of BD and has been shown to exacerbate IRI, although the role of BD and complement in VCA and the role it plays in IRI and VCA rejection has not been studied. Methods BD was induced in Balb/c donors, and the VCA perfused prior to graft procurement with UW solution supplemented with or without CR2-Crry, a C3 convertase complement inhibitor that binds at sites of complement activation, such as that induced on the endothelium by induction of BD. Following perfusion, donor VCAs were cold stored for 6 hours before transplantation into C57BL/6 recipients. Donor VCAs from living donors (LD) were also procured and stored. Analyses included CR2-Crry graft binding, complement activation, toxicity, injury/inflammation, graft gene expression and survival. Results Compared to LD VCAs, BD donor VCAs had exacerbated IRI and rejected earlier. Following pretransplant in-situ perfusion of the donor graft, CR2-Crry bound within the graft and was retained post-transplantation. CR2-Crry treatment significantly reduced complement deposition, inflammation and IRI as compared to vehicle-treated BD donors. Treatment of BD donor VCAs with CR2-Crry led to an injury profile not dissimilar to that seen in recipients of LD VCAs. Conclusion Pre-coating a VCA with CR2-Crry in a clinically relevant treatment paradigm provides localized, and therefore minimally immunosuppressive, protection from the complement-mediated effects of BD induced exacerbated IRI.
Collapse
Affiliation(s)
- Biao Lei
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - M. Mahdi Sleiman
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Qi Cheng
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Surgery, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Institute of Organ Transplantation, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenxiao Tu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Surgery, Hepatic and Vascular Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zhu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Surgery, Hepatic and Vascular Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Martin Goddard
- Pathology Department, Papworth Hospital NHS Trust, Cambridge, United Kingdom
| | - Paulo N. Martins
- UMass Memorial Medical Center, Department of Surgery, Transplant Division, University of Massachusetts, Worcester, MA, United States
| | - Logan Langerude
- Division of Pulmonary Medicine, University of Florida, Gainesville, FL, United States
| | - Satish Nadig
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Microbiology and Immunology, Charleston, SC, United States
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Microbiology and Immunology, Charleston, SC, United States
- Ralph H. Johnson VA Medical Center, Charleston, SC, United States
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Division of Pulmonary Medicine, University of Florida, Gainesville, FL, United States
- Department of Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Microbiology and Immunology, Charleston, SC, United States
| |
Collapse
|
13
|
Li C, Patel K, Tu Z, Yang X, Kulik L, Alawieh A, Allen P, Cheng Q, Wallace C, Kilkenny J, Kwon J, Gibney B, Cantu E, Sharma A, Pipkin M, Machuca T, Emtiazjoo A, Goddard M, Holers VM, Nadig S, Christie J, Tomlinson S, Atkinson C. A novel injury site-natural antibody targeted complement inhibitor protects against lung transplant injury. Am J Transplant 2021; 21:2067-2078. [PMID: 33210808 PMCID: PMC8246004 DOI: 10.1111/ajt.16404] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 01/25/2023]
Abstract
Complement is known to play a role in ischemia and reperfusion injury (IRI). A general paradigm is that complement is activated by self-reactive natural IgM antibodies (nAbs), after they engage postischemic neoepitopes. However, a role for nAbs in lung transplantation (LTx) has not been explored. Using mouse models of LTx, we investigated the role of two postischemic neoepitopes, modified annexin IV (B4) and a subset of phospholipids (C2), in LTx. Antibody deficient Rag1-/- recipient mice were protected from LTx IRI. Reconstitution with either B4 or C2nAb restored IRI, with C2 significantly more effective than B4 nAb. Based on these information, we developed/characterized a novel complement inhibitor composed of single-chain antibody (scFv) derived from the C2 nAb linked to Crry (C2scFv-Crry), a murine inhibitor of C3 activation. Using an allogeneic LTx, in which recipients contain a full nAb repertoire, C2scFv-Crry targeted to the LTx, inhibited IRI, and delayed acute rejection. Finally, we demonstrate the expression of the C2 neoepitope in human donor lungs, highlighting the translational potential of this approach.
Collapse
Affiliation(s)
- Changhai Li
- The Hepatic Surgery Centre at Tongji Hospital, Tongji Medical College, HUST, Wuhan, China
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
- Department of Microbiology and Immunology, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
| | - Kunal Patel
- Department of Microbiology and Immunology, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
- Department of Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
| | - Zhenxiao Tu
- Department of Microbiology and Immunology, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
- Department of Surgery, Hepatic and Vascular Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofeng Yang
- Department of Microbiology and Immunology, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
| | - Liudmila Kulik
- Department of Medicine and Immunology, University of Colorado Denver, Aurora, Colorado, USA
| | - Ali Alawieh
- Department of Microbiology and Immunology, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
| | - Patterson Allen
- Department of Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
| | - Qi Cheng
- The Hepatic Surgery Centre at Tongji Hospital, Tongji Medical College, HUST, Wuhan, China
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Caroline Wallace
- Department of Microbiology and Immunology, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
- Department of Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
| | - Jane Kilkenny
- Department of Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
| | - Jennie Kwon
- Department of Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
| | - Barry Gibney
- Department of Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
| | - Edward Cantu
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
| | - Ashish Sharma
- Department of Surgery, University of Florida, Gainesville, Florida, USA
| | - Mauricio Pipkin
- Division of Thoracic and Cardiovascular Surgery, University of Florida, Gainesville, Florida, USA
| | - Tiago Machuca
- Division of Thoracic and Cardiovascular Surgery, University of Florida, Gainesville, Florida, USA
| | - Amir Emtiazjoo
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, Florida, USA
| | - Martin Goddard
- Pathology Department, Papworth Hospital, NHS Trust, Papworth Everard, Cambridge, UK
| | - V Michael Holers
- Department of Medicine and Immunology, University of Colorado Denver, Aurora, Colorado, USA
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Satish Nadig
- Department of Microbiology and Immunology, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
- Department of Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
- South Carolina Investigators in Transplantation, Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jason Christie
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
- Department of Surgery, University of Florida, Gainesville, Florida, USA
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina, USA
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
- Department of Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Microbiology and Immunology, Charleston, South Carolina, USA
- South Carolina Investigators in Transplantation, Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
14
|
Heart Transplantation From Brain Dead Donors: A Systematic Review of Animal Models. Transplantation 2021; 104:2272-2289. [PMID: 32150037 DOI: 10.1097/tp.0000000000003217] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Despite advances in mechanical circulatory devices and pharmacologic therapies, heart transplantation (HTx) is the definitive and most effective therapy for an important proportion of qualifying patients with end-stage heart failure. However, the demand for donor hearts significantly outweighs the supply. Hearts are sourced from donors following brain death, which exposes donor hearts to substantial pathophysiological perturbations that can influence heart transplant success and recipient survival. Although significant advances in recipient selection, donor and HTx recipient management, immunosuppression, and pretransplant mechanical circulatory support have been achieved, primary graft dysfunction after cardiac transplantation continues to be an important cause of morbidity and mortality. Animal models, when appropriate, can guide/inform medical practice, and fill gaps in knowledge that are unattainable in clinical settings. Consequently, we performed a systematic review of existing animal models that incorporate donor brain death and subsequent HTx and assessed studies for scientific rigor and clinical relevance. Following literature screening via the U.S National Library of Medicine bibliographic database (MEDLINE) and Embase, 29 studies were assessed. Analysis of included studies identified marked heterogeneity in animal models of donor brain death coupled to HTx, with few research groups worldwide identified as utilizing these models. General reporting of important determinants of heart transplant success was mixed, and assessment of posttransplant cardiac function was limited to an invasive technique (pressure-volume analysis), which is limitedly applied in clinical settings. This review highlights translational challenges between available animal models and clinical heart transplant settings that are potentially hindering advancement of this field of investigation.
Collapse
|
15
|
Hu ZG, Zhou Y, Lin CJ, Yuan GD, He SQ. Emerging recognition of the complement system in hepatic ischemia/reperfusion injury, liver regeneration and recovery (Review). Exp Ther Med 2021; 21:223. [PMID: 33603832 PMCID: PMC7851628 DOI: 10.3892/etm.2021.9654] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatic ischemia/reperfusion injury (IRI) is a result of the ischemic cascade and may occur in the settings of liver trauma, resection and transplantation. Components of the complement system have been indicated to be mediators of hepatic IRI and regulators of liver regeneration. As such, their potential to mediate both beneficial and harmful effects render them key targets for therapy. In the present study, the mechanisms of complement mediating hepatic IRI were discussed with a focus on the different functions of complement in hepatic injury and liver recovery, and an explanation for this apparent paradox is provided, i.e. that the complement products C3a and C5a have an important role in liver damage; however, C3a and C5a are also necessary for liver regeneration. Furthermore, situated at the end of the complement activation cascade, the membrane attack complex is crucial in hepatic IRI and inhibiting the complex with a site-targeted murine complement inhibitor, complement receptor 2-CD59, may improve liver regeneration after partial hepatectomy, even when hepatectomy is combined with ischemia and reperfusion.
Collapse
Affiliation(s)
- Zhi-Gao Hu
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yi Zhou
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Cheng-Jie Lin
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Guan-Dou Yuan
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Song-Qing He
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
16
|
Kulkarni HS, Ramphal K, Ma L, Brown M, Oyster M, Speckhart KN, Takahashi T, Byers DE, Porteous MK, Kalman L, Hachem RR, Rushefski M, McPhatter J, Cano M, Kreisel D, Scavuzzo M, Mittler B, Cantu E, Pilely K, Garred P, Christie JD, Atkinson JP, Gelman AE, Diamond JM. Local complement activation is associated with primary graft dysfunction after lung transplantation. JCI Insight 2020; 5:138358. [PMID: 32750037 PMCID: PMC7526453 DOI: 10.1172/jci.insight.138358] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/29/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The complement system plays a key role in host defense but is activated by ischemia/reperfusion injury (IRI). Primary graft dysfunction (PGD) is a form of acute lung injury occurring predominantly due to IRI, which worsens survival after lung transplantation (LTx). Local complement activation is associated with acute lung injury, but whether it is more reflective of allograft injury compared with systemic activation remains unclear. We proposed that local complement activation would help identify those who develop PGD after LTx. We also aimed to identify which complement activation pathways are associated with PGD. METHODS We performed a multicenter cohort study at the University of Pennsylvania and Washington University School of Medicine. Bronchoalveolar lavage (BAL) and plasma specimens were obtained from recipients within 24 hours after LTx. PGD was scored based on the consensus definition. Complement activation products and components of each arm of the complement cascade were measured using ELISA. RESULTS In both cohorts, sC4d and sC5b-9 levels were increased in BAL of subjects with PGD compared with those without PGD. Subjects with PGD also had higher C1q, C2, C4, and C4b, compared with subjects without PGD, suggesting classical and lectin pathway involvement. Ba levels were higher in subjects with PGD, suggesting alternative pathway activation. Among lectin pathway–specific components, MBL and FCN-3 had a moderate-to-strong correlation with the terminal complement complex in the BAL but not in the plasma. CONCLUSION Complement activation fragments are detected in the BAL within 24 hours after LTx. Components of all 3 pathways are locally increased in subjects with PGD. Our findings create a precedent for investigating complement-targeted therapeutics to mitigate PGD. FUNDING This research was supported by the NIH, American Lung Association, Children’s Discovery Institute, Robert Wood Johnson Foundation, Cystic Fibrosis Foundation, Barnes-Jewish Hospital Foundation, Danish Heart Foundation, Danish Research Foundation of Independent Research, Svend Andersen Research Foundation, and Novo Nordisk Research Foundation. Substantial differences between local and systemic complement activation in lung transplant recipients who develop primary graft dysfunction are identified in two independent cohorts.
Collapse
Affiliation(s)
- Hrishikesh S Kulkarni
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kristy Ramphal
- Department of Medicine, Perlman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lina Ma
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Melanie Brown
- Department of Medicine, Perlman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michelle Oyster
- Department of Medicine, Perlman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kaitlyn N Speckhart
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tsuyoshi Takahashi
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Derek E Byers
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mary K Porteous
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Laurel Kalman
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ramsey R Hachem
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Melanie Rushefski
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ja'Nia McPhatter
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marlene Cano
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Brigitte Mittler
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Edward Cantu
- Department of Surgery, Perlman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katrine Pilely
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jason D Christie
- Department of Medicine, Perlman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John P Atkinson
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrew E Gelman
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joshua M Diamond
- Department of Medicine, Perlman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
17
|
Frye CC, Bery AI, Kreisel D, Kulkarni HS. Sterile inflammation in thoracic transplantation. Cell Mol Life Sci 2020; 78:581-601. [PMID: 32803398 DOI: 10.1007/s00018-020-03615-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/20/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
The life-saving benefits of organ transplantation can be thwarted by allograft dysfunction due to both infectious and sterile inflammation post-surgery. Sterile inflammation can occur after necrotic cell death due to the release of endogenous ligands [such as damage-associated molecular patterns (DAMPs) and alarmins], which perpetuate inflammation and ongoing cellular injury via various signaling cascades. Ischemia-reperfusion injury (IRI) is a significant contributor to sterile inflammation after organ transplantation and is associated with detrimental short- and long-term outcomes. While the vicious cycle of sterile inflammation and cellular injury is remarkably consistent amongst different organs and even species, we have begun understanding its mechanistic basis only over the last few decades. This understanding has resulted in the developments of novel, yet non-specific therapies for mitigating IRI-induced graft damage, albeit with moderate results. Thus, further understanding of the mechanisms underlying sterile inflammation after transplantation is critical for identifying personalized therapies to prevent or interrupt this vicious cycle and mitigating allograft dysfunction. In this review, we identify common and distinct pathways of post-transplant sterile inflammation across both heart and lung transplantation that can potentially be targeted.
Collapse
Affiliation(s)
- C Corbin Frye
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Amit I Bery
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, 4523 Clayton Avenue, Campus Box 8052, St. Louis, MO, 63110, USA.
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hrishikesh S Kulkarni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, 4523 Clayton Avenue, Campus Box 8052, St. Louis, MO, 63110, USA
| |
Collapse
|
18
|
Khan MA, Shamma T, Kazmi S, Altuhami A, Ahmed HA, Assiri AM, Broering DC. Hypoxia-induced complement dysregulation is associated with microvascular impairments in mouse tracheal transplants. J Transl Med 2020; 18:147. [PMID: 32234039 PMCID: PMC7110829 DOI: 10.1186/s12967-020-02305-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 03/16/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Complement Regulatory Proteins (CRPs), especially CD55 primarily negate complement factor 3-mediated injuries and maintain tissue homeostasis during complement cascade activation. Complement activation and regulation during alloimmune inflammation contribute to allograft injury and therefore we proposed to investigate a crucial pathological link between vascular expression of CD55, active-C3, T cell immunity and associated microvascular tissue injuries during allograft rejection. METHODS Balb/c→C57BL/6 allografts were examined for microvascular deposition of CD55, C3d, T cells, and associated tissue microvascular impairments during rejection in mouse orthotopic tracheal transplantation. RESULTS Our findings demonstrated that hypoxia-induced early activation of HIF-1α favors a cell-mediated inflammation (CD4+, CD8+, and associated proinflammatory cytokines, IL-2 and TNF-α), which proportionally triggers the downregulation of CRP-CD55, and thereby augments the uncontrolled release of active-C3, and Caspase-3 deposition on CD31+ graft vascular endothelial cells. These molecular changes are pathologically associated with microvascular deterioration (low tissue O2 and Blood flow) and subsequent airway epithelial injuries of rejecting allografts as compared to non-rejecting syngrafts. CONCLUSION Together, these findings establish a pathological correlation between complement dysregulation, T cell immunity, and microvascular associated injuries during alloimmune inflammation in transplantation.
Collapse
Affiliation(s)
- Mohammad Afzal Khan
- Organ Transplant Research Section, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia.
| | - Talal Shamma
- Organ Transplant Research Section, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Shadab Kazmi
- Organ Transplant Research Section, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Abdullah Altuhami
- Organ Transplant Research Section, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Hala Abdalrahman Ahmed
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Abdullah Mohammed Assiri
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia.,Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Kingdom of Saudi Arabia
| | - Dieter Clemens Broering
- Organ Transplant Research Section, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
19
|
Liu DQ, Chen SP, Sun J, Wang XM, Chen N, Zhou YQ, Tian YK, Ye DW. Berberine protects against ischemia-reperfusion injury: A review of evidence from animal models and clinical studies. Pharmacol Res 2019; 148:104385. [PMID: 31400402 DOI: 10.1016/j.phrs.2019.104385] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/27/2019] [Accepted: 07/31/2019] [Indexed: 12/15/2022]
Abstract
Ischemia-reperfusion (I/R) injury is accompanied with high morbidity and mortality and has seriously negative social and economic influences. Unfortunately, few effective therapeutic strategies are available to improve its outcome. Berberine is a natural medicine possessing multiple beneficial biological activities. Emerging evidence indicates that berberine has potential protective effects against I/R injury in brain, heart, kidney, liver, intestine and testis. However, up-to-date review focusing on the beneficial role of berberine against I/R injury is not yet available. In this paper, results from animal models and clinical studies are concisely presented and its mechanisms are discussed. We found that berberine ameliorates I/R injury in animal models via its anti-oxidant, anti-apoptotic and anti-inflammatory effects. Moreover, berberine also attenuates I/R injury by suppressing endoplasmic reticulum stress and promoting autophagy. Additionally, regulation of periphery immune system may also contributes to the beneficial effect of berberine against I/R injury. Although clinical evidence is limited, the current studies indicate that berberine may attenuate I/R injury via inhibiting excessive inflammatory response in patients. Collectively, berberine might be used as an alternative therapeutic strategy for the management of I/R injury.
Collapse
Affiliation(s)
- Dai-Qiang Liu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu-Ping Chen
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Sun
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Mei Wang
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nan Chen
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Qun Zhou
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yu-Ke Tian
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Da-Wei Ye
- Cancer Center, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
20
|
Hu C, Zhao L, Wu D, Li L. Modulating autophagy in mesenchymal stem cells effectively protects against hypoxia- or ischemia-induced injury. Stem Cell Res Ther 2019; 10:120. [PMID: 30995935 PMCID: PMC6471960 DOI: 10.1186/s13287-019-1225-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In mammals, a basal level of autophagy, a self-eating cellular process, degrades cytosolic proteins and subcellular organelles in lysosomes to provide energy, recycles the cytoplasmic components, and regenerates cellular building blocks; thus, autophagy maintains cellular and tissue homeostasis in all eukaryotic cells. In general, adaptive autophagy increases when cells confront stressful conditions to improve the survival rate of the cells, while destructive autophagy is activated when the cellular stress is not manageable and elicits the regenerative capacity. Hypoxia-reoxygenation (H/R) injury and ischemia-reperfusion (I/R) injury initiate excessive autophagy and endoplasmic reticulum (ER) stress and consequently induce a string of damage in mammalian tissues or organs. Mesenchymal stem cell (MSC)-based therapy has yielded promising results in repairing H/R- or I/R-induced injury in various tissues. However, MSC transplantation in vivo must overcome the barriers including the low survival rate of transplanted stem cells, limited targeting capacity, and low grafting potency; therefore, much effort is needed to increase the survival and activity of MSCs in vivo. Modulating autophagy regulates the stemness and the anti-oxidative stress, anti-apoptosis, and pro-survival capacity of MSCs and can be applied to MSC-based therapy for repairing H/R- or I/R-induced cellular or tissue injury.
Collapse
Affiliation(s)
- Chenxia Hu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lingfei Zhao
- Kidney Disease Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Daxian Wu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lanjuan Li
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
21
|
Ma Y, Liu Y, Zhang Z, Yang GY. Significance of Complement System in Ischemic Stroke: A Comprehensive Review. Aging Dis 2019; 10:429-462. [PMID: 31011487 PMCID: PMC6457046 DOI: 10.14336/ad.2019.0119] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/19/2019] [Indexed: 12/14/2022] Open
Abstract
The complement system is an essential part of innate immunity, typically conferring protection via eliminating pathogens and accumulating debris. However, the defensive function of the complement system can exacerbate immune, inflammatory, and degenerative responses in various pathological conditions. Cumulative evidence indicates that the complement system plays a critical role in the pathogenesis of ischemic brain injury, as the depletion of certain complement components or the inhibition of complement activation could reduce ischemic brain injury. Although multiple candidates modulating or inhibiting complement activation show massive potential for the treatment of ischemic stroke, the clinical availability of complement inhibitors remains limited. The complement system is also involved in neural plasticity and neurogenesis during cerebral ischemia. Thus, unexpected side effects could be induced if the systemic complement system is inhibited. In this review, we highlighted the recent concepts and discoveries of the roles of different kinds of complement components, such as C3a, C5a, and their receptors, in both normal brain physiology and the pathophysiology of brain ischemia. In addition, we comprehensively reviewed the current development of complement-targeted therapy for ischemic stroke and discussed the challenges of bringing these therapies into the clinic. The design of future experiments was also discussed to better characterize the role of complement in both tissue injury and recovery after cerebral ischemia. More studies are needed to elucidate the molecular and cellular mechanisms of how complement components exert their functions in different stages of ischemic stroke to optimize the intervention of targeting the complement system.
Collapse
Affiliation(s)
- Yuanyuan Ma
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yanqun Liu
- 3Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhijun Zhang
- 2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
22
|
Khan MA, Shamma T. Complement factor and T-cell interactions during alloimmune inflammation in transplantation. J Leukoc Biol 2018; 105:681-694. [PMID: 30536904 DOI: 10.1002/jlb.5ru0718-288r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/25/2018] [Accepted: 11/21/2018] [Indexed: 02/06/2023] Open
Abstract
Complement factor and T-cell signaling during an effective alloimmune response plays a key role in transplant-associated injury, which leads to the progression of chronic rejection (CR). During an alloimmune response, activated complement factors (C3a and C5a) bind to their corresponding receptors (C3aR and C5aR) on a number of lymphocytes, including T-regulatory cells (Tregs), and these cell-molecular interactions have been vital to modulate an effective immune response to/from Th1-effector cell and Treg activities, which result in massive inflammation, microvascular impairments, and fibrotic remodeling. Involvement of the complement-mediated cell signaling during transplantation signifies a crucial role of complement components as a key therapeutic switch to regulate ongoing inflammatory state, and further to avoid the progression of CR of the transplanted organ. This review highlights the role of complement-T cell interactions, and how these interactions shunt the effector immune response during alloimmune inflammation in transplantation, which could be a novel therapeutic tool to protect a transplanted organ and avoid progression of CR.
Collapse
Affiliation(s)
- Mohammad Afzal Khan
- Organ Transplant Research Section, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Talal Shamma
- Organ Transplant Research Section, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
23
|
Ansari MA, Iqubal A, Ekbbal R, Haque SE. Effects of nimodipine, vinpocetine and their combination on isoproterenol-induced myocardial infarction in rats. Biomed Pharmacother 2018; 109:1372-1380. [PMID: 30551388 DOI: 10.1016/j.biopha.2018.10.199] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/22/2018] [Accepted: 10/31/2018] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Myocardial infarction (MI) remains a major cause of morbidity and mortality worldwide. Nimodipine is a calcium (Ca2+) channel blocker as well as a PDE1 inhibitor and primarily used in subarachnoid haemorrhage (SAH) due to its blood-brain barrier crossing property. Nimodipine and vinpocetine inhibit the degradation of phosphodiester bond which increases cGMP and cAMP levels causing vasodilation. MATERIAL AND METHODS We have divided rats randomly into Group I - Vehicle control; Group II - Toxic control (ISO 85 mg/kg, i.p.); Group III, IV and V - Nimodipine (5, 10 and 15 mg/kg, i.p. respectively) with ISO; Group VI- Nimodipine (15 mg/kg) alone; Group VII - Nimodipine + Vinpocetine (10 mg/kg + 10 mg/kg) with ISO; Group VIII - Nimodipine + Vinpocetine (10 mg/kg + 10 mg/kg) alone; Group IX- Diltiazem (25 mg/kg, p.o) with ISO; Group X- Diltiazem (25 mg/kg) alone and Group XI- Vinpocetine (10 mg/kg, p.o.) with ISO for 7 days. After 24 h of the last dose, haemodynamics were assessed then animals were sacrificed and biochemical, histopathological and ultrastructural changes were measured. RESULTS Treatment with ISO significantly deviated the haemodynamic parameters (HR, SAP, DAP and MAP), biochemical parameters (CK-MB, LDH, SGOT, cGMP and Troponin-T) and antioxidant markers (TBARS, SOD, CAT, GSH, GPx, GST and GR). Haemotoxylin and eosin staining of the cardiac tissue and ultrastructural study also indicated significant myocardial damage. Pretreatment with nimodipine (10 and 15 mg/kg, i.p), vinpocetine (10 mg/kg, p.o) and their combination significantly restored the antioxidant status, haemodynamic profile, cellular architecture and ultrastructural changes in the heart. CONCLUSION Nimodipine and vinpocetine both showed cardioprotection when given alone. However, their combination showed better restoration in terms of oxidative stress, cardiac membrane damage, haemodynamics, histopathology and ultrastructural changes.
Collapse
Affiliation(s)
- Mohd Asif Ansari
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi, 110062, India
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi, 110062, India
| | - Rustam Ekbbal
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi, 110062, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
24
|
Cheng Q, Patel K, Lei B, Rucker L, Allen DP, Zhu P, Vasu C, Martins PN, Goddard M, Nadig SN, Atkinson C. Donor pretreatment with nebulized complement C3a receptor antagonist mitigates brain-death induced immunological injury post-lung transplant. Am J Transplant 2018; 18:2417-2428. [PMID: 29504277 PMCID: PMC6123303 DOI: 10.1111/ajt.14717] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 02/16/2018] [Accepted: 02/19/2018] [Indexed: 01/25/2023]
Abstract
Donor brain death (BD) is an inherent part of lung transplantation (LTx) and a key contributor to ischemia-reperfusion injury (IRI). Complement activation occurs as a consequence of BD in other solid organ Tx and exacerbates IRI, but the role of complement in LTx has not been investigated. Here, we investigate the utility of delivering nebulized C3a receptor antagonist (C3aRA) pretransplant to BD donor lungs in order to reduce post-LTx IRI. BD was induced in Balb/c donors, and lungs nebulized with C3aRA or vehicle 30 minutes prior to lung procurement. Lungs were then cold stored for 18 hours before transplantation into C57Bl/6 recipients. Donor lungs from living donors (LD) were removed and similarly stored. At 6 hours and 5 days post-LTx, recipients of BD donor lungs had exacerbated IRI and acute rejection (AR), respectively, compared to recipients receiving LD lungs, as determined by increased histopathological injury, immune cells, and cytokine levels. A single pretransplant nebulized dose of C3aRA to the donor significantly reduced IRI as compared to vehicle-treated BD donors, and returned IRI and AR grades to that seen following LD LTx. These data demonstrate a role for complement inhibition in the amelioration of IRI post-LTx in the context of donor BD.
Collapse
Affiliation(s)
- Qi Cheng
- Institute of Organ Transplantation, Department of Surgery, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan,430030, China,Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA,Department of Surgery, Division of Transplant, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Kunal Patel
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA,Department of Surgery, Division of Transplant, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Biao Lei
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Lindsay Rucker
- Department of Surgery, Division of Transplant, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - D. Patterson Allen
- Department of Surgery, Division of Transplant, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Peng Zhu
- Institute of Organ Transplantation, Department of Surgery, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan,430030, China,Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA,Department of Surgery, Division of Transplant, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Chentha Vasu
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Paulo N. Martins
- University of Massachusetts, UMass Memorial Medical Center, Department of Surgery, Transplant Division, Worcester, MA 01655, USA
| | - Martin Goddard
- Pathology Department, Papworth Hospital NHS Trust, Papworth Everard, Cambridge, England, CB3 8RE
| | - Satish N. Nadig
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA,Department of Surgery, Division of Transplant, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA,South Carolina Investigators in Transplantation (SCIT), Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA,Department of Surgery, Division of Transplant, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA,South Carolina Investigators in Transplantation (SCIT), Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA,Address for Correspondence. Dr Carl Atkinson, PhD. Department of Microbiology and Immunology, and Surgery. Medical University of South Carolina, Lee Patterson Allen Transplant Immunobiology Laboratory, Basic Science Department, 173 Ashley Avenue, Charleston, SC 29425 USA. Tel: 1-843-792-1716. Fax: 1-843-792-2464.
| |
Collapse
|
25
|
Tissue-targeted complement therapeutics. Mol Immunol 2018; 102:120-128. [PMID: 30220307 DOI: 10.1016/j.molimm.2018.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 02/07/2023]
Abstract
Complement activation contributes to the pathogenesis of numerous inflammatory and autoimmune diseases. Therapeutic complement inhibitors have proven effective in several of these diseases and have now entered clinical use. Complement activation has multiple different biologic effects, however, and the currently available drugs can have undesirable side-effects, such as an increased risk of infection. Several different complement inhibitors have been developed that bind to target molecules, thereby concentrating the drug at a specific anatomic site. This approach appears to be both more effective than untargeted drugs and to have fewer side effects. In this article we review different targeting strategies that have been developed and the evidence supporting the use and benefits of targeted drugs.
Collapse
|
26
|
Annamalai B, Parsons N, Belhaj M, Brandon C, Potts J, Rohrer B. Encapsulated Cell Technology-Based Delivery of a Complement Inhibitor Reduces Choroidal Neovascularization in a Mouse Model. Transl Vis Sci Technol 2018; 7:3. [PMID: 29576927 PMCID: PMC5846441 DOI: 10.1167/tvst.7.2.3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/19/2017] [Indexed: 12/28/2022] Open
Abstract
Purpose Age-related macular degeneration (AMD) is a slowly progressing disease, and risk appears to be tied to an overactive complement system. We have previously demonstrated that mouse choroidal neovascularization (CNV) and smoke-induced ocular pathology can be reduced with an alternative pathway (AP) inhibitor fusion protein consisting of a complement receptor-2 fragment linked to the inhibitory domain of factor H (CR2-fH) when delivered systemically. Here we developed an experimental approach with genetically engineered encapsulated ARPE-19 cells to produce CR2-fH intravitreally. Methods ARPE-19 cells were generated to stably express CR2 or CR2-fH, microencapsulated using sodium alginate, and injected intravitreally into 2-month-old C57BL/6J mice. CNV was induced using argon laser photocoagulation 4 weeks postinjection. Presence of capsules and progression of CNV was analyzed using optical coherence tomography. Bioavailability of CR2-fH was evaluated in retina sections by immunohistochemistry, and efficacy as an AP inhibitor by C3a ELISA. Results Secretion of CR2-fH or CR2 from encapsulated ARPE-19 cells was confirmed. An efficacious concentration of CR2-fH capsules to reduce CNV was identified. Bioavailability studies showed that CR2-fH was present in capsules and retinas of injected mice, and reduced CNV-associated ocular C3a production. Conclusions These findings indicate that the AP inhibitor CR2-fH, when generated intravitreally, can reduce CNV in mouse. Translational Relevance Encapsulated ARPE-19 cells secreting CR2-fH or perhaps other antiangiogenic or prosurvival factors might be useful as a potential therapeutic tool to treat age-related macular degeneration.
Collapse
Affiliation(s)
| | - Nathaniel Parsons
- Departments of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA
| | - Marwa Belhaj
- Department of Cell Biology, University of South Carolina, Columbia, SC, USA
| | - Carlene Brandon
- Departments of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA
| | - Jay Potts
- Department of Cell Biology, University of South Carolina, Columbia, SC, USA
| | - Bärbel Rohrer
- Departments of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA.,Neurosciences, Division of Research, Medical University of South Carolina, Charleston, SC, USA.,Ralph H. Johnson VA Medical Center, Division of Research, Charleston, SC, USA
| |
Collapse
|
27
|
Effects of C1 inhibitor on endothelial cell activation in a rat hind limb ischemia-reperfusion injury model. J Vasc Surg 2018; 68:209S-221S.e2. [PMID: 29395422 DOI: 10.1016/j.jvs.2017.10.072] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/11/2017] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Ischemia-reperfusion (I/R) injury is a major clinical problem linked to vascular surgery. Currently, no drugs to prevent or to treat I/R injury are approved for clinical use. C1 inhibitor (C1 INH) is known to reduce activation of the plasma cascade systems that are involved in the pathophysiologic process of I/R injury. The aim of this study was therefore to investigate the effect of C1 INH on complement deposition and endothelial cell activation in a rat model of hind limb I/R injury. METHODS Male Wistar rats (wild type, bred at the central animal facility, University of Bern), weighing 250 to 320 g, were used. The rats underwent 2-hour ischemia and 24-hour reperfusion by unilateral clamping of the femoral artery and additional use of a tourniquet. Five groups were divided according to intravenous treatment 5 minutes before ischemia: 50 IU/kg C1 INH (n = 5); 100 IU/kg C1 INH (n = 7); vehicle control (n = 5); nontreated control (n = 7); and normal, healthy control without intervention (n = 4). At the end, muscle edema, tissue viability, and histologic features were assessed. Deposition of immunoglobulin M, C1r, C4d, and fibrin and expression of plasminogen activator inhibitor 1, heparan sulfate (HS), E-selectin, and vascular cell adhesion molecule 1 were evaluated by fluorescence staining. In addition, high-mobility group box 1 protein was measured in plasma. RESULTS Edema formation was reduced by C1 INH at two dosages, mirrored by improved histologic injury scores and preserved muscle viability. Deposition of immunoglobulin M, C4d, and fibrin was significantly decreased by 100 IU/kg C1 INH compared with nontreated controls. Pretreatment with 100 IU/kg C1 INH also significantly reduced HS shedding and expression of plasminogen activator inhibitor 1 as well as plasma levels of high-mobility group box 1 protein. CONCLUSIONS Pretreatment with both 50 and 100 IU/kg C1 INH attenuated reperfusion injury of rat hind limbs. Pretreatment with 100 IU/kg also preserved the endothelial HS layer as well as the natural, profibrinolytic phenotype of the endothelium. Prevention of endothelial cell activation by C1 INH may therefore be a promising strategy to prevent I/R injury in the clinical setting of peripheral vascular diseases and elective surgery on extremities.
Collapse
|
28
|
Khan V, Sharma S, Bhandari U, Ali SM, Haque SE. Raspberry ketone protects against isoproterenol-induced myocardial infarction in rats. Life Sci 2017; 194:205-212. [PMID: 29225109 DOI: 10.1016/j.lfs.2017.12.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/28/2017] [Accepted: 12/06/2017] [Indexed: 12/12/2022]
Abstract
AIM The cardioprotective role of raspberry ketone (RK) against isoproterenol (ISO)-induced myocardial infarction (MI) in rats was assessed. MATERIALS AND METHODS Rats were randomly divided into Group I - Vehicle control; Group II - Toxic control ISO (85mg/kg, s.c.); Group III, IV and V - RK (50, 100 and 200mg/kg, respectively) with ISO; Group VI- RK (200mg/kg) alone; Group VII - Propranolol (10mg/kg) with ISO; and Group VIII - Propranolol (10mg/kg) alone. After twenty-four hours of the last dose, animals were sacrificed and creatine kinase-MB, lactate dehydrogenase, total cholesterol, triglycerides, high-density-lipoprotein, low-density-lipoprotein, very-low-density-lipoprotein, malondialdehyde, reduced glutathione, superoxide dismutase, catalase, Na+, K+-ATPase, nitric oxide, histopathological and immunohistochemical analysis (tumor necrosis factor-α and inducible nitric oxide synthase) were performed. KEY FINDINGS Treatment with ISO significantly deviated the biochemical parameters from the normal levels, which were considerably restored by RK at 100 and 200mg/kg doses. 50mg/kg dose, however, did not demonstrate any significant cardioprotective action. The histopathological and immunohistochemical analysis further substantiated these findings. SIGNIFICANCE Our study showed a dose-dependent reduction in oxidative stress, inflammation and dyslipidemia by RK in ISO-intoxicated rats, which signifies that RK from the European red raspberry plant might be a valuable entity for the management of MI.
Collapse
Affiliation(s)
- Vasim Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi 110062, India
| | - Sumit Sharma
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi 110062, India
| | - Uma Bhandari
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi 110062, India
| | - Syed Mansoor Ali
- Department of Biotechnology, Jamia Milia Islamia, New Delhi 110025, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
29
|
Chun N, Haddadin AS, Liu J, Hou Y, Wong KA, Lee D, Rushbrook JI, Gulaya K, Hines R, Hollis T, Nistal Nuno B, Mangi AA, Hashim S, Pekna M, Catalfamo A, Chin HY, Patel F, Rayala S, Shevde K, Heeger PS, Zhang M. Activation of complement factor B contributes to murine and human myocardial ischemia/reperfusion injury. PLoS One 2017; 12:e0179450. [PMID: 28662037 PMCID: PMC5491012 DOI: 10.1371/journal.pone.0179450] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/29/2017] [Indexed: 12/28/2022] Open
Abstract
The pathophysiology of myocardial injury that results from cardiac ischemia and reperfusion (I/R) is incompletely understood. Experimental evidence from murine models indicates that innate immune mechanisms including complement activation via the classical and lectin pathways are crucial. Whether factor B (fB), a component of the alternative complement pathway required for amplification of complement cascade activation, participates in the pathophysiology of myocardial I/R injury has not been addressed. We induced regional myocardial I/R injury by transient coronary ligation in WT C57BL/6 mice, a manipulation that resulted in marked myocardial necrosis associated with activation of fB protein and myocardial deposition of C3 activation products. In contrast, in fB-/- mice, the same procedure resulted in significantly reduced myocardial necrosis (% ventricular tissue necrotic; fB-/- mice, 20 ± 4%; WT mice, 45 ± 3%; P < 0.05) and diminished deposition of C3 activation products in the myocardial tissue (fB-/- mice, 0 ± 0%; WT mice, 31 ± 6%; P<0.05). Reconstitution of fB-/- mice with WT serum followed by cardiac I/R restored the myocardial necrosis and activated C3 deposition in the myocardium. In translational human studies we measured levels of activated fB (Bb) in intracoronary blood samples obtained during cardio-pulmonary bypass surgery before and after aortic cross clamping (AXCL), during which global heart ischemia was induced. Intracoronary Bb increased immediately after AXCL, and the levels were directly correlated with peripheral blood levels of cardiac troponin I, an established biomarker of myocardial necrosis (Spearman coefficient = 0.465, P < 0.01). Taken together, our results support the conclusion that circulating fB is a crucial pathophysiological amplifier of I/R-induced, complement-dependent myocardial necrosis and identify fB as a potential therapeutic target for prevention of human myocardial I/R injury.
Collapse
Affiliation(s)
- Nicholas Chun
- Nephrology Division, Department of Medicine and Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ala S. Haddadin
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Junying Liu
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Yunfang Hou
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Karen A. Wong
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Daniel Lee
- Department of Surgery, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Julie I. Rushbrook
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Karan Gulaya
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Roberta Hines
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Tamika Hollis
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Beatriz Nistal Nuno
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Abeel A. Mangi
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Sabet Hashim
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Marcela Pekna
- Department of Medical Chemistry and Cell Biology, Göteborg University, Göteborg, Sweden
| | - Amy Catalfamo
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Hsiao-ying Chin
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Foramben Patel
- Department of Biomedical Sciences, Long Island University, Brookville, New York, United States of America
| | - Sravani Rayala
- Department of Biomedical Sciences, Long Island University, Brookville, New York, United States of America
| | - Ketan Shevde
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Peter S. Heeger
- Nephrology Division, Department of Medicine and Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ming Zhang
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
- Department of Cell Biology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| |
Collapse
|
30
|
Targeted Complement Inhibition Protects Vascularized Composite Allografts From Acute Graft Injury and Prolongs Graft Survival When Combined With Subtherapeutic Cyclosporine A Therapy. Transplantation 2017; 101:e75-e85. [PMID: 28045880 DOI: 10.1097/tp.0000000000001625] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Recipients of vascularized composite allografts require aggressive and lifelong immunosuppression, and because the surgery is usually performed in nonlife-threatening situations, the development of strategies to minimize immunosuppression is especially pertinent for this procedure. We investigated how complement affects acute graft injury, alloimmunity, and immunosuppressive therapy. METHODS Vascularized composite allografts were transplanted from Balb/C to C57BL/6 mice that were complement deficient (C3 or double C3a Receptor (R)/C5aR), or treated with a targeted complement inhibitor (CR2-Crry). Allografts were analyzed for acute inflammation and injury, subacute T cell response, and survival in the absence and presence of cyclosporine A (CsA) therapy. RESULTS Allografts in C3-deficient or CR2-Crry-treated recipients were protected from skin and muscle ischemia-reperfusion injury (IRI). C3aR/C5aR-deficient recipients were more modestly protected. IgM and C3d colocalized within allografts from wild type and C3aR/C5aR-deficient recipients indicating IgM-mediated complement activation, and C3d deposition was almost absent in allografts from C3-deficient and CR2-Crry-treated recipients. Inflammatory cell infiltration and P-selectin expression was also significantly reduced in C3-deficient and CR2-Crry-treated recipients. Acute treatment with CR2-Crry or with 3 mg/kg per day CsA modestly, but significantly increased median allograft survival from 5.8 to 7.4 and 7.2 days, respectively. However, combined acute CR2-Crry treatment and CsA therapy increased mean graft survival to 17.2 days. Protection was associated with significantly reduced T cell infiltration of allografts and Tc1 cells in recipient spleens. CONCLUSIONS Complement-mediated IRI augments graft allogenicity, and appropriate complement inhibition ameliorates IRI, decreases alloimmune priming and allows more immune-sparing CsA dosing.
Collapse
|
31
|
Qin L, Li G, Kirkiles-Smith N, Clark P, Fang C, Wang Y, Yu ZX, Devore D, Tellides G, Pober JS, Jane-wit D. Complement C5 Inhibition Reduces T Cell-Mediated Allograft Vasculopathy Caused by Both Alloantibody and Ischemia Reperfusion Injury in Humanized Mice. Am J Transplant 2016; 16:2865-2876. [PMID: 27104811 PMCID: PMC5075274 DOI: 10.1111/ajt.13834] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 04/10/2016] [Accepted: 04/14/2016] [Indexed: 01/25/2023]
Abstract
Allograft vasculopathy (AV) is characterized by diffuse stenoses in the vasculature of solid organ transplants. Previously, we developed two humanized models showing that alloantibody and ischemia reperfusion injury (IRI) exacerbated T cell-mediated AV in human arterial xenografts in vivo. Herein we examined a causal role for terminal complement activation in both settings. IRI, in contrast to alloantibody, elicited widespread membrane attack complex (MAC) assembly throughout the vessel wall. Both alloantibody and IRI caused early (24 h) and robust endothelial cell (EC) activation localized to regions of intimal MAC deposition, indicated by increases in nuclear factor kappa B (NF-κB)-inducing kinase, an MAC-dependent activator of noncanonical NF-kB, VCAM-1 expression and Gr-1+ neutrophil infiltration. Endothelial cell activation by alloantibody was inhibited by antimouse C5 mAb, but not by anti-C5a mAb or by control mAb, implicating MAC as the primary target of anti-C5 mAb. Antimouse C5 mAb significantly reduced alloantibody- and IRI-enhanced T cell infiltration and AV-like changes, including neointimal hyperplasia as well as intraluminal thrombosis in a subset of IRI-treated arterial grafts. These results indicate that increased AV lesion formation in response to either alloantibody or IRI is dependent on complement C5 activation and, accordingly, inhibition of this pathway may attenuate AV.
Collapse
Affiliation(s)
- Lingfeng Qin
- Department of Surgery, Yale School of Medicine, New Haven, CT 06519
| | - Guangxin Li
- Department of Surgery, Yale School of Medicine, New Haven, CT 06519
| | | | - Pamela Clark
- Immunobiology, Yale School of Medicine, New Haven, CT 06519
| | - Caodi Fang
- Immunobiology, Yale School of Medicine, New Haven, CT 06519
| | - Yi Wang
- Alexion Pharmaceuticals Inc., 100 College St, New Haven, CT 06511
| | - Zhao-Xue Yu
- Alexion Pharmaceuticals Inc., 100 College St, New Haven, CT 06511
| | - Denise Devore
- Alexion Pharmaceuticals Inc., 100 College St, New Haven, CT 06511
| | - George Tellides
- Department of Surgery, Yale School of Medicine, New Haven, CT 06519
| | - Jordan S Pober
- Immunobiology, Yale School of Medicine, New Haven, CT 06519
| | - Dan Jane-wit
- Cardiovascular Medicine, Yale School of Medicine, New Haven, CT 06519
| |
Collapse
|
32
|
Woodell A, Jones BW, Williamson T, Schnabolk G, Tomlinson S, Atkinson C, Rohrer B. A Targeted Inhibitor of the Alternative Complement Pathway Accelerates Recovery From Smoke-Induced Ocular Injury. Invest Ophthalmol Vis Sci 2016; 57:1728-37. [PMID: 27064393 PMCID: PMC4829088 DOI: 10.1167/iovs.15-18471] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose Morphologic and genetic evidence exists that an overactive complement system driven by the complement alternative pathway (AP) is involved in pathogenesis of age-related macular degeneration (AMD). Smoking is the only modifiable risk factor for AMD. As we have shown that smoke-related ocular pathology can be prevented in mice that lack an essential activator of AP, we ask here whether this pathology can be reversed by increasing inhibition in AP. Methods Mice were exposed to either cigarette smoke (CS) or filtered air (6 hours/day, 5 days/week, 6 months). Smoke-exposed animals were then treated with the AP inhibitor (CR2-fH) or vehicle control (PBS) for the following 3 months. Spatial frequency and contrast sensitivity were assessed by optokinetic response paradigms at 6 and 9 months; additional readouts included assessment of retinal morphology by electron microscopy (EM) and gene expression analysis by quantitative RT-PCR. Results The CS mice treated with CR2-fH showed significant improvement in contrast threshold compared to PBS-treated mice, whereas spatial frequency was unaffected by CS or pharmacologic intervention. Treatment with CR2-fH in CS animals reversed thinning of the retina observed in PBS-treated mice as analyzed by spectral-domain optical coherence tomography, and reversed most morphologic changes in RPE and Bruch's membrane seen in CS animals by EM. Conclusions Taken together, these findings suggest that AP inhibitors not only prevent, but have the potential to accelerate the clearance of complement-mediated ocular injury. Improving our understanding of the regulation of the AP is paramount to developing novel treatment approaches for AMD.
Collapse
Affiliation(s)
- Alex Woodell
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Bryan W Jones
- Moran Eye Center, University of Utah, Salt Lake City, Utah, United States
| | - Tucker Williamson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Gloriane Schnabolk
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, South Carolina, United States
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States 4Research Service, Ralph H. Johnson VA Medical Center, Charleston, South Carolina, United States
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States 5Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Bärbel Rohrer
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, United States 4Research Service, Ralph H. Johnson VA Medical Center, Charleston, South Carolina, United States 6Department of Ophthalmology, Medical University o
| |
Collapse
|
33
|
Montero RM, Sacks SH, Smith RA. Complement-here, there and everywhere, but what about the transplanted organ? Semin Immunol 2016; 28:250-9. [PMID: 27179705 DOI: 10.1016/j.smim.2016.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/20/2016] [Accepted: 04/26/2016] [Indexed: 12/15/2022]
Abstract
The part of the innate immune system that communicates and effectively primes the adaptive immune system was termed "complement" by Ehrlich to reflect its complementarity to antibodies having previously been described as "alexine" (i.e protective component of serum) by Buchner and Bordet. It has been established that complement is not solely produced systemically but may have origin in different tissues where it can influence organ specific functions that may affect the outcome of transplanted organs. This review looks at the role of complement in particular to kidney transplantation. We look at current literature to determine whether blockade of the peripheral or central compartments of complement production may prevent ischaemic reperfusion injury or rejection in the transplanted organ. We also review new therapeutics that have been developed to inhibit components of the complement cascade with varying degrees of success leading to an increase in our understanding of the multiple triggers of this complex system. In addition, we consider whether biomarkers in this field are effective markers of disease or treatment.
Collapse
Affiliation(s)
- R M Montero
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, NIHR Comprehensive Biomedical Research Centre, King's College London, Guy's & St Thomas' NHS Foundation Trust, United Kingdom
| | - S H Sacks
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, NIHR Comprehensive Biomedical Research Centre, King's College London, Guy's & St Thomas' NHS Foundation Trust, United Kingdom.
| | - R A Smith
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, NIHR Comprehensive Biomedical Research Centre, King's College London, Guy's & St Thomas' NHS Foundation Trust, United Kingdom
| |
Collapse
|
34
|
Khan MA, Hsu JL, Assiri AM, Broering DC. Targeted complement inhibition and microvasculature in transplants: a therapeutic perspective. Clin Exp Immunol 2015; 183:175-86. [PMID: 26404106 DOI: 10.1111/cei.12713] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2015] [Indexed: 12/18/2022] Open
Abstract
Active complement mediators play a key role in graft-versus-host diseases, but little attention has been given to the angiogenic balance and complement modulation during allograft acceptance. The complement cascade releases the powerful proinflammatory mediators C3a and C5a anaphylatoxins, C3b, C5b opsonins and terminal membrane attack complex into tissues, which are deleterious if unchecked. Blocking complement mediators has been considered to be a promising approach in the modern drug discovery plan, and a significant number of therapeutic alternatives have been developed to dampen complement activation and protect host cells. Numerous immune cells, especially macrophages, develop both anaphylatoxin and opsonin receptors on their cell surface and their binding affects the macrophage phenotype and their angiogenic properties. This review discusses the mechanism that complement contributes to angiogenic injury, and the development of future therapeutic targets by antagonizing activated complement mediators to preserve microvasculature in rejecting the transplanted organ.
Collapse
Affiliation(s)
- M A Khan
- Organ Transplant Centre, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - J L Hsu
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - A M Assiri
- Organ Transplant Centre, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - D C Broering
- Organ Transplant Centre, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
35
|
Abstract
Complement is a key component of immunity with crucial inflammatory and opsonic properties; inappropriate activation of complement triggers or exacerbates inflammatory disease. Complement dysregulation is a core feature of some diseases and contributes to pathology in many others. Approved agents have been developed for and are highly effective in some orphan applications, but their progress to use in more common diseases has been slow. Numerous challenges, such as target concentration or high turnover, limit the efficacy of these agents in humans. Numerous novel agents targeting different parts of the complement system in different ways are now emerging from pre-clinical studies and are entering Phase I/II trials; these agents bring the potential for more-effective and more-specific anti-complement therapies in disease. Other agents, both biologic and small molecule, are in Phase II or III trials for both rare and common diseases — administration routes include localized (for example, intravitreal) and systemic routes. There is an urgent need to develop biomarkers and imaging methods that enable monitoring of the effects and efficacy of anti-complement agents.
The complement cascade, a key regulator of innate immunity, is a rich source of potential therapeutic targets for diseases including autoimmune, inflammatory and degenerative disorders. Morgan and Harris discuss the progress made in modulating the complement system and the existing challenges, including dosing, localization of the drug to the target and how to interfere with protein–protein interactions. The complement system is a key innate immune defence against infection and an important driver of inflammation; however, these very properties can also cause harm. Inappropriate or uncontrolled activation of complement can cause local and/or systemic inflammation, tissue damage and disease. Complement provides numerous options for drug development as it is a proteolytic cascade that involves nine specific proteases, unique multimolecular activation and lytic complexes, an arsenal of natural inhibitors, and numerous receptors that bind to activation fragments. Drug design is facilitated by the increasingly detailed structural understanding of the molecules involved in the complement system. Only two anti-complement drugs are currently on the market, but many more are being developed for diseases that include infectious, inflammatory, degenerative, traumatic and neoplastic disorders. In this Review, we describe the history, current landscape and future directions for anti-complement therapies.
Collapse
|
36
|
Sharif‐Paghaleh E, Yap ML, Meader LL, Chuamsaamarkkee K, Kampmeier F, Badar A, Smith RA, Sacks S, Mullen GE. Noninvasive Imaging of Activated Complement in Ischemia-Reperfusion Injury Post-Cardiac Transplant. Am J Transplant 2015; 15:2483-90. [PMID: 25906673 PMCID: PMC4654255 DOI: 10.1111/ajt.13299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 02/24/2015] [Accepted: 02/28/2015] [Indexed: 01/25/2023]
Abstract
Ischemia-reperfusion injury (IRI) is inevitable in solid organ transplantation, due to the transplanted organ being ischemic for prolonged periods prior to transplantation followed by reperfusion. The complement molecule C3 is present in the circulation and is also synthesized by tissue parenchyma in early response to IRI and the final stable fragment of activated C3, C3d, can be detected on injured tissue for several days post-IRI. Complement activation post-IRI was monitored noninvasively by single photon emission computed tomography (SPECT) and CT using (99m) Tc-recombinant complement receptor 2 ((99m) Tc-rCR2) in murine models of cardiac transplantation following the induction of IRI and compared to (99m) Tc-rCR2 in C3(-/-) mice or with the irrelevant protein (99m) Tc-prostate-specific membrane antigen antibody fragment (PSMA). Significant uptake with (99m) Tc-rCR2 was observed as compared to C3(-/-) or (99m) Tc-PSMA. In addition, the transplanted heart to muscle ratio of (99m) Tc-rCR2 was significantly higher than (99m) Tc-PSMA or C3(-/-) . The results were confirmed by histology and autoradiography. (99m) Tc-rCR2 can be used for noninvasive detection of activated complement and in future may be used to quantify the severity of transplant damage due to complement activation postreperfusion.
Collapse
Affiliation(s)
- E. Sharif‐Paghaleh
- Division of Imaging and Biomedical EngineeringSchool of MedicineKing's College LondonLondonEngland,MRC Centre for TransplantationKing's College LondonLondonEngland,Department of ImmunologyFaculty of MedicineTehran University of Medical SciencesTehranIran
| | - M. L. Yap
- Division of Imaging and Biomedical EngineeringSchool of MedicineKing's College LondonLondonEngland
| | - L. L. Meader
- MRC Centre for TransplantationKing's College LondonLondonEngland
| | - K. Chuamsaamarkkee
- Division of Imaging and Biomedical EngineeringSchool of MedicineKing's College LondonLondonEngland
| | - F. Kampmeier
- Division of Imaging and Biomedical EngineeringSchool of MedicineKing's College LondonLondonEngland
| | - A. Badar
- Division of Imaging and Biomedical EngineeringSchool of MedicineKing's College LondonLondonEngland
| | - R. A. Smith
- MRC Centre for TransplantationKing's College LondonLondonEngland
| | - S. Sacks
- MRC Centre for TransplantationKing's College LondonLondonEngland
| | - G. E. Mullen
- Division of Imaging and Biomedical EngineeringSchool of MedicineKing's College LondonLondonEngland,MRC Centre for TransplantationKing's College LondonLondonEngland
| |
Collapse
|
37
|
Nadig SN, Dixit SK, Levey N, Esckilsen S, Miller K, Dennis W, Atkinson C, Broome AM. Immunosuppressive nano-therapeutic micelles downregulate endothelial cell inflammation and immunogenicity. RSC Adv 2015; 5:43552-43562. [PMID: 26167278 PMCID: PMC4494678 DOI: 10.1039/c5ra04057d] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In this study, we developed a stable, nontoxic novel micelle nanoparticle to attenuate responses of endothelial cell (EC) inflammation when subjected to oxidative stress, such as observed in organ transplantation. Targeted Rapamycin Micelles (TRaM) were synthesized using PEG-PE-amine and N-palmitoyl homocysteine (PHC) with further tailoring of the micelle using targeting peptides (cRGD) and labeling with far-red fluorescent dye for tracking during cellular uptake studies. Our results revealed that the TRaM was approximately 10 nm in diameter and underwent successful internalization in Human Umbilical Vein EC (HUVEC) lines. Uptake efficiency of TRaM nanoparticles was improved with the addition of a targeting moiety. In addition, our TRaM therapy was able to downregulate both mouse cardiac endothelial cell (MCEC) and HUVEC production and release of the pro-inflammatory cytokines, IL-6 and IL-8 in normal oxygen tension and hypoxic conditions. We were also able to demonstrate a dose-dependent uptake of TRaM therapy into biologic tissues ex vivo. Taken together, these data demonstrate the feasibility of targeted drug delivery in transplantation, which has the potential for conferring local immunosuppressive effects without systemic consequences while also dampening endothelial cell injury responses.
Collapse
Affiliation(s)
- Satish N Nadig
- Department of Surgery, Division of Transplant, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA. ; Tel: 01 843 792 8596;
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA. ; Tel: 01 843 792 1716;
- South Carolina Investigators in Transplantation (SCIT), Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA. ; Tel: 01 843 792 3553;
| | - Suraj K Dixit
- Department of Radiology & Radiological Science, Medical University of South Carolina, 68 President Street MSC 120, Charleston, SC 29425, USA. ; Tel: 01 843 876 2481;
- Center for Biomedical Imaging (CBI), Medical University of South Carolina, 68 President Street MSC 120, Charleston, SC 29425, USA. ; Tel: 01 843 876 2481;
| | - Natalie Levey
- Department of Surgery, Division of Transplant, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA. ; Tel: 01 843 792 8596;
| | - Scott Esckilsen
- Department of Surgery, Division of Transplant, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA. ; Tel: 01 843 792 8596;
| | - Kayla Miller
- Department of Radiology & Radiological Science, Medical University of South Carolina, 68 President Street MSC 120, Charleston, SC 29425, USA. ; Tel: 01 843 876 2481;
- Center for Biomedical Imaging (CBI), Medical University of South Carolina, 68 President Street MSC 120, Charleston, SC 29425, USA. ; Tel: 01 843 876 2481;
| | - William Dennis
- Department of Surgery, Division of Transplant, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA. ; Tel: 01 843 792 8596;
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA. ; Tel: 01 843 792 1716;
- South Carolina Investigators in Transplantation (SCIT), Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA. ; Tel: 01 843 792 3553;
| | - Ann-Marie Broome
- Department of Radiology & Radiological Science, Medical University of South Carolina, 68 President Street MSC 120, Charleston, SC 29425, USA. ; Tel: 01 843 876 2481;
- Center for Biomedical Imaging (CBI), Medical University of South Carolina, 68 President Street MSC 120, Charleston, SC 29425, USA. ; Tel: 01 843 876 2481;
- South Carolina Investigators in Transplantation (SCIT), Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA. ; Tel: 01 843 792 3553;
| |
Collapse
|
38
|
Atkinson C, Qiao F, Yang X, Zhu P, Reaves N, Kulik L, Goddard M, Holers VM, Tomlinson S. Targeting pathogenic postischemic self-recognition by natural IgM to protect against posttransplantation cardiac reperfusion injury. Circulation 2015; 131:1171-80. [PMID: 25825397 DOI: 10.1161/circulationaha.114.010482] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Natural IgM antibodies represent a class of innate pattern recognition receptors that recognize danger-associated molecular patterns expressed on stressed or dying cells. They play important roles in tissue homeostasis by disposing of prenecrotic cells and suppressing inflammation. However, ischemic insult leads to a pathogenic level of IgM binding and complement activation, resulting in inflammation and injury. We investigate the role of self-reactive IgM in the unique setting of transplantation where the donor organ undergoes both cold and warm ischemia and global ischemic insult. METHODS AND RESULTS By transplanting hearts from wild-type donor mice into antibody-deficient mice reconstituted with specific self-reactive IgM monoclonal antibodies, we identified neoepitopes expressed after transplantation and demonstrated a key role for IgM recognition of these epitopes in graft injury. With this information, we developed and characterized a therapeutic strategy that exploited the postischemia recognition system of natural antibodies. On the basis of neoepitope identification, we constructed an anti-annexin IV single-chain antibody (scFv) and an scFv linked to Crry, an inhibitor of C3 activation (scFv-Crry). In an allograft transplantation model in which recipients contain a full natural antibody repertoire, both constructs blocked graft IgM binding and complement activation and significantly reduced graft inflammation and injury. Furthermore, scFv-Crry specifically targeted to the transplanted heart and, unlike complement deficiency, did not affect immunity to infection, an important consideration for immunosuppressed transplant recipients. CONCLUSIONS We identified pathophysiologically important epitopes expressed within the heart after transplantation and described a novel translatable strategy for targeted complement inhibition that has several advantages over currently available approaches.
Collapse
Affiliation(s)
- Carl Atkinson
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Fei Qiao
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Xiaofeng Yang
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Peng Zhu
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Nicholas Reaves
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Liudmila Kulik
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Martin Goddard
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - V Michael Holers
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Stephen Tomlinson
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.).
| |
Collapse
|
39
|
Di Paolo NC, Baldwin LK, Irons EE, Papayannopoulou T, Tomlinson S, Shayakhmetov DM. IL-1α and complement cooperate in triggering local neutrophilic inflammation in response to adenovirus and eliminating virus-containing cells. PLoS Pathog 2014; 10:e1004035. [PMID: 24651866 PMCID: PMC3961377 DOI: 10.1371/journal.ppat.1004035] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 02/14/2014] [Indexed: 12/13/2022] Open
Abstract
Inflammation is a highly coordinated host response to infection, injury, or cell stress. In most instances, the inflammatory response is pro-survival and is aimed at restoring physiological tissue homeostasis and eliminating invading pathogens, although exuberant inflammation can lead to tissue damage and death. Intravascular injection of adenovirus (Ad) results in virus accumulation in resident tissue macrophages that trigger activation of CXCL1 and CXCL2 chemokines via the IL-1α-IL-1RI signaling pathway. However, the mechanistic role and functional significance of this pathway in orchestrating cellular inflammatory responses to the virus in vivo remain unclear. Resident metallophilic macrophages expressing macrophage receptor with collagenous structure (MARCO+) in the splenic marginal zone (MZ) play the principal role in trapping Ad from the blood. Here we show that intravascular Ad administration leads to the rapid recruitment of Ly-6G+7/4+ polymorphonuclear leukocytes (PMNs) in the splenic MZ, the anatomical compartment that remains free of PMNs when these cells are purged from the bone marrow via a non-inflammatory stimulus. Furthermore, PMN recruitment in the splenic MZ resulted in elimination of virus-containing cells. IL-1α-IL-1RI signaling is only partially responsible for PMN recruitment in the MZ and requires CXCR2, but not CXCR1 signaling. We further found reduced recruitment of PMNs in the splenic MZ in complement C3-deficient mice, and that pre-treatment of IL-1α-deficient, but not wild-type mice, with complement inhibitor CR2-Crry (inhibits all complement pathways at C3 activation) or CR2-fH (inhibits only the alternative complement activation pathway) prior to Ad infection, abrogates PMN recruitment to the MZ and prevents elimination of MARCO+ macrophages from the spleen. Collectively, our study reveals a non-redundant role of the molecular factors of innate immunity – the chemokine-activating IL-1α-IL-1RI-CXCR2 axis and complement – in orchestrating local inflammation and functional cooperation of PMNs and resident macrophages in the splenic MZ, which collectively contribute to limiting disseminated pathogen spread via elimination of virus-containing cells. Adenovirus (Ad) induces a potent activation of pro-inflammatory cytokines and chemokines upon interaction with tissue macrophages in vivo. However, critical factors affecting cellular inflammatory responses to Ad and their functional significance remain unclear. Here we show that in the model of disseminated infection, intravenous Ad administration leads to a rapid release of pro-inflammatory Ly-6G+7/4+ leukocytes (PMNs) from the bone marrow into the blood. PMNs enter into peripheral tissues and, in the case of spleen, are accumulated in proximity to the virus-containing MARCO+ macrophages within the splenic marginal zone (MZ). Mechanistic dissection of molecular queues that guide PMN migration reveals that CXCL1 and CXCL2 chemokines are only partially responsible for CXCR2-dependent PMN recruitment into the splenic MZ. We further found that complement cooperates with IL-1α-IL-1RI-CXCR2 signaling pathways in recruitment of PMNs to the splenic MZ, which results in elimination of virus-containing MARCO+ macrophages from the spleen. Administration of complement-blocking CR2-Crry or CR2-fH proteins into IL-1α-deficient, but not wild-type, mice prevents PMN accumulation in the splenic MZ and elimination of virus-containing macrophages from the spleen. Our study defines the functional significance of molecular and cellular host defense mechanisms that cooperate in eliminating virus-containing cells in the model of acute disseminated Ad infection.
Collapse
Affiliation(s)
- Nelson C. Di Paolo
- Lowance Center for Human Immunology, Departments of Pediatrics and Medicine, Emory University School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Lisa K. Baldwin
- Division of Medical Genetics Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Eric E. Irons
- Division of Medical Genetics Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Thalia Papayannopoulou
- Division of Hematology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States of America
| | - Dmitry M. Shayakhmetov
- Lowance Center for Human Immunology, Departments of Pediatrics and Medicine, Emory University School of Medicine, Emory University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
40
|
Liu S, Xing T, Sheng T, Yang S, Huang L, Peng Z, Sun X. The reduction rate of serum C3 following liver transplantation is an effective predictor of non-anastomotic strictures. Hepatol Int 2014. [DOI: 10.1007/s12072-014-9524-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
41
|
Physiological and therapeutic complement regulators in kidney transplantation. Curr Opin Organ Transplant 2013; 18:421-9. [PMID: 23838647 DOI: 10.1097/mot.0b013e32836370ce] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW This review will summarize the key contribution of complement regulators in the immune response to an allograft. RECENT FINDINGS Over the past 10 years, compelling evidences have been accumulated in support of a critical role of complement in the pathological phenomena related to organ transplantation. In addition to recurrence of complement-mediated disease after graft, complement is involved in situations as diverse as brain death induced tissue damages, ischaemia-reperfusion and antibody-mediated rejections. This complement activation is counterbalanced by various regulatory mechanisms. SUMMARY We discuss the role of physiological and therapeutic complement regulators that are designed to overcome the impact of complement overactivation with the aim of improving long-term transplant outcomes. We will focus primarily on renal allograft, but the discussed mechanisms take place to a different degree in any kind of organ transplantation.
Collapse
|
42
|
Zaferani A, Talsma D, Richter MKS, Daha MR, Navis GJ, Seelen MA, van den Born J. Heparin/heparan sulphate interactions with complement--a possible target for reduction of renal function loss? Nephrol Dial Transplant 2013; 29:515-22. [PMID: 23880790 DOI: 10.1093/ndt/gft243] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Current management of end-stage renal failure is based on renal replacement therapy by dialysis or transplantation. Increased occurrence of renal failure in both native and transplanted kidneys indicates a need for novel therapies to stop or limit the progression of the disease. Acute kidney injury and proteinuria are major risk factors in the development of renal failure. In this regard, innate immunity plays an important role in the pathogenesis of renal diseases in both native and transplanted kidneys. The complement system is a major humoral part of innate defense. Next to the well-known complement activators, quite a number of the complement factors react with proteoglycans (PGs) both on cellular membranes and in the extracellular compartment. Therefore, these interactions might serve as targets for intervention. In this review, the current knowledge of interactions between PGs and complement is reviewed, and additionally the options for interference in the progression of renal disease are discussed.
Collapse
Affiliation(s)
- Azadeh Zaferani
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
43
|
Woodell A, Coughlin B, Kunchithapautham K, Casey S, Williamson T, Ferrell WD, Atkinson C, Jones BW, Rohrer B. Alternative complement pathway deficiency ameliorates chronic smoke-induced functional and morphological ocular injury. PLoS One 2013; 8:e67894. [PMID: 23825688 PMCID: PMC3692454 DOI: 10.1371/journal.pone.0067894] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 05/23/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Age-related macular degeneration (AMD), a complex disease involving genetic variants and environmental insults, is among the leading causes of blindness in Western populations. Genetic and histologic evidence implicate the complement system in AMD pathogenesis; and smoking is the major environmental risk factor associated with increased disease risk. Although previous studies have demonstrated that cigarette smoke exposure (CE) causes retinal pigment epithelium (RPE) defects in mice, and smoking leads to complement activation in patients, it is unknown whether complement activation is causative in the development of CE pathology; and if so, which complement pathway is required. METHODS Mice were exposed to cigarette smoke or clean, filtered air for 6 months. The effects of CE were analyzed in wildtype (WT) mice or mice without a functional complement alternative pathway (AP; CFB(-/-) ) using molecular, histological, electrophysiological, and behavioral outcomes. RESULTS CE in WT mice exhibited a significant reduction in function of both rods and cones as determined by electroretinography and contrast sensitivity measurements, concomitant with a thinning of the nuclear layers as measured by SD-OCT imaging and histology. Gene expression analyses suggested that alterations in both photoreceptors and RPE/choroid might contribute to the observed loss of function, and visualization of complement C3d deposition implies the RPE/Bruch's membrane (BrM) complex as the target of AP activity. RPE/BrM alterations include an increase in mitochondrial size concomitant with an apical shift in mitochondrial distribution within the RPE and a thickening of BrM. CFB(-/-) mice were protected from developing these CE-mediated alterations. CONCLUSIONS Taken together, these findings provide clear evidence that ocular pathology generated in CE mice is dependent on complement activation and requires the AP. Identifying animal models with RPE/BrM damage and verifying which aspects of pathology are dependent upon complement activation is essential for developing novel complement-based treatment approaches for the treatment of AMD.
Collapse
Affiliation(s)
- Alex Woodell
- Division of Research, Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Beth Coughlin
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Kannan Kunchithapautham
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Sarah Casey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Tucker Williamson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - W. Drew Ferrell
- Moran Eye Center, University of Utah, Salt Lake City, Utah, United States of America
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Bryan W. Jones
- Moran Eye Center, University of Utah, Salt Lake City, Utah, United States of America
| | - Bärbel Rohrer
- Division of Research, Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, South Carolina, United States of America
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Organ transplantation and other major surgeries are impacted by ischemia-reperfusion injury (IRI). Mesenchymal stromal cells (MSCs) recently became an attractive alternative therapeutic tool to combat IRI. The present review highlights the effects of MSCs in the preclinical animal models of IRI and clinical trials, and explains their potential modes of action based on the pathophysiological IRI cascade. RECENT FINDINGS The application of MSCs in animal models of IRI show anti-inflammatory and anti-apoptotic effects, particularly for damage to the kidneys, heart and lungs. The mechanism of MSC action remains unclear, but may involve paracrine factors which could include the transfer of microvesicles, RNA or mitochondria. Although few clinical trials have reached completion, adverse effects appear minimal. SUMMARY MSCs show promise in protecting against IRI-induced damage. They appear to help recovery mainly by affecting the levels of inflammation and apoptosis during the organ repair process. In addition, they may mediate immunomodulatory effects on the innate and adaptive immune processes triggered during reperfusion and reduce fibrosis. Success in preclinical animal models has led to the initiation of ongoing clinical trials.
Collapse
|
45
|
Atkinson C, Floerchinger B, Qiao F, Casey S, Williamson T, Moseley E, Stoica S, Goddard M, Ge X, Tullius SG, Tomlinson S. Donor brain death exacerbates complement-dependent ischemia/reperfusion injury in transplanted hearts. Circulation 2013; 127:1290-9. [PMID: 23443736 DOI: 10.1161/circulationaha.112.000784] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Brain death (BD) can immunologically prime the donor organ and is thought to lead to exacerbated ischemia/reperfusion injury after transplantation. Using a newly developed mouse model of BD, we investigated the effect of donor BD on posttransplantation cardiac ischemia/reperfusion injury. We further investigated the therapeutic effect of a targeted complement inhibitor in recipients of BD donor hearts and addressed the clinical relevance of these studies by analyzing human heart biopsies from BD and domino (living) donors. METHODS AND RESULTS Hearts from living or BD donor C57BL/6 mice were transplanted into C57BL/6 or BALB/c recipients. Recipient mice were treated with the complement inhibitor CR2-Crry or vehicle control (n=6). Isografts were analyzed 48 hours after transplantation for injury, inflammation, and complement deposition, and allografts were monitored for graft survival. Human cardiac biopsies were analyzed for complement deposition and inflammatory cell infiltration. In the murine model, donor BD exacerbated ischemia/reperfusion injury and graft rejection, as demonstrated by increased myocardial injury, serum cardiac troponin, cellular infiltration, complement deposition, inflammatory chemokine and cytokine levels, and by decreased graft survival. CR2-Crry treatment of recipients significantly reduced all measured outcomes in grafts from both BD and living donors compared with controls. Analysis of human samples documented the relevance of our experimental findings and revealed exacerbated complement deposition and inflammation in grafts from BD donors compared with grafts from living donors. CONCLUSIONS BD exacerbates posttransplantation cardiac ischemia/reperfusion injury in mice and humans and decreases survival of mouse allografts. Furthermore, targeted complement inhibition in recipient mice ameliorates BD-exacerbated ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
CR2-mediated targeting of complement inhibitors: bench-to-bedside using a novel strategy for site-specific complement modulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 735:137-54. [PMID: 23402024 DOI: 10.1007/978-1-4614-4118-2_9] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent approval of the first human complement pathway-directed therapeutics, along with high-profile genetic association studies, has catalyzed renewed biopharmaceutical interest in developing drugs that modulate the complement system. Substantial challenges remain, however, that must be overcome before widespread application of complement inhibitors in inflammatory and autoimmune diseases becomes possible. Among these challenges are the following: (1) defining the complement pathways and effector mechanisms that cause tissue injury in humans and determining whether the relative importance of each varies by disease, (2) blocking or modulating, using traditional small molecule or biologic approaches, the function of complement proteins whose circulating levels are very high and whose turnover rates are relatively rapid, especially in the setting of acute and chronic autoimmune diseases, and (3) avoiding infectious complications or impairment of other important physiological functions of complement when using systemically active complement-blocking agents. This chapter will review data that address these challenges to therapeutic development, with a focus on the development of a novel strategy of blocking specific complement pathways by targeting inhibitors using a recombinant portion of the human complement receptor type 2 (CR2/CD21) which specifically targets to sites of local complement C3 activation where C3 fragments are covalently fixed. Recently, the first of these CR2-targeted proteins has entered human phase I studies in the human disease paroxysmal nocturnal hemoglobinuria. The results of murine translational studies using CR2-targeted inhibitors strongly suggest that a guiding principle going forward in complement therapeutic development may well be to focus on developing strategies to modulate the pathway as precisely as possible by physically localizing therapeutic inhibitory effects.
Collapse
|
47
|
Abstract
The complement cascade is a major contributor to the innate immune response. It has now been well accepted that complement plays a critical role in hyperacute rejection and acute antibody-mediated rejection of transplanted organ. There is also increasing evidence that complement proteins contribute to the pathogenesis of organ ischemia-reperfusion injury, and even to cell-mediated rejection. Furthermore, the chemoattractants C3a and C5a and the terminal membrane attack complex that are generated by complement activation can directly or indirectly mediate tissue injury and trigger adaptive immune responses. Here, we review recent findings concerning the role of complement in graft ischemia-reperfusion injury, antibody-mediated rejection and accommodation, and cell-mediated rejection. We also discuss the current status of complement intervention therapies in clinical transplantation and describe potential new therapeutic strategies for clinical application.
Collapse
Affiliation(s)
- Gang Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | |
Collapse
|
48
|
Brennan FH, Anderson AJ, Taylor SM, Woodruff TM, Ruitenberg MJ. Complement activation in the injured central nervous system: another dual-edged sword? J Neuroinflammation 2012; 9:137. [PMID: 22721265 PMCID: PMC3464784 DOI: 10.1186/1742-2094-9-137] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 06/21/2012] [Indexed: 11/28/2022] Open
Abstract
The complement system, a major component of the innate immune system, is becoming increasingly recognised as a key participant in physiology and disease. The awareness that immunological mediators support various aspects of both normal central nervous system (CNS) function and pathology has led to a renaissance of complement research in neuroscience. Various studies have revealed particularly novel findings on the wide-ranging involvement of complement in neural development, synapse elimination and maturation of neural networks, as well as the progression of pathology in a range of chronic neurodegenerative disorders, and more recently, neurotraumatic events, where rapid disruption of neuronal homeostasis potently triggers complement activation. The purpose of this review is to summarise recent findings on complement activation and acquired brain or spinal cord injury, i.e. ischaemic-reperfusion injury or stroke, traumatic brain injury (TBI) and spinal cord injury (SCI), highlighting the potential for complement-targeted therapeutics to alleviate the devastating consequences of these neurological conditions.
Collapse
Affiliation(s)
- Faith H Brennan
- The University of Queensland, School of Biomedical Sciences, St Lucia, Brisbane, QLD 4072, Australia
| | | | | | | | | |
Collapse
|
49
|
Abstract
The complement system is a key element of the innate immune system, and the production of complement components can be divided into central (hepatic) and peripheral compartments. Essential complement components such as C3 are produced in both of these compartments, but until recently the functional relevance of the peripheral synthesis of complement was unclear. Here, we review recent findings showing that local peripheral synthesis of complement in a transplanted organ is required for the immediate response of the donor organ to tissue stress and for priming alloreactive T cells that can mediate transplant rejection. We also discuss recent insights into the role of complement in antibody-mediated rejection, and we examine how new treatment strategies that take into account the separation of central and peripheral production of complement are expected to make a difference to transplant outcome.
Collapse
|