1
|
Ali AE, Becker RC. The foundation for investigating factor XI as a target for inhibition in human cardiovascular disease. J Thromb Thrombolysis 2024; 57:1283-1296. [PMID: 38662114 PMCID: PMC11645312 DOI: 10.1007/s11239-024-02985-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/13/2024] [Indexed: 04/26/2024]
Abstract
Anticoagulant therapy is a mainstay in the management of patients with cardiovascular disease and related conditions characterized by a heightened risk for thrombosis. Acute coronary syndrome, chronic coronary syndrome, ischemic stroke, and atrial fibrillation are the most common. In addition to their proclivity for thrombosis, each of these four conditions is also characterized by local and systemic inflammation, endothelial/endocardial injury and dysfunction, oxidative stress, impaired tissue-level reparative capabilities, and immune dysregulation that plays a critical role in linking molecular events, environmental triggers, and phenotypic expressions. Knowing that cardiovascular disease and thrombosis are complex and dynamic, can the scientific community identify a common pathway or specific point of interface susceptible to pharmacological inhibition or alteration that is likely to be safe and effective? The contact factors of coagulation may represent the proverbial "sweet spot" and are worthy of investigation. The following review provides a summary of the fundamental biochemistry of factor XI, its biological activity in thrombosis, inflammation, and angiogenesis, new targeting drugs, and a pragmatic approach to managing hemostatic requirements in clinical trials and possibly day-to-day patient care in the future.
Collapse
Affiliation(s)
- Ahmed E Ali
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Richard C Becker
- Department of Internal Medicine, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA.
| |
Collapse
|
2
|
Kasbe R, Tripathy AS, Wani MR, Mullick J. Elevated Complement Activation Fragments and C1q-Binding Circulating Immune Complexes in Varied Phases of Chikungunya Virus Infection. Curr Microbiol 2024; 81:242. [PMID: 38913141 DOI: 10.1007/s00284-024-03732-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 05/06/2024] [Indexed: 06/25/2024]
Abstract
Chikungunya virus (CHIKV) is a causative agent of a disease continuum, ranging from an acute transient chikungunya fever to chronic incapacitating viral arthralgia. The interaction between anti-CHIKV antibodies and the complement system has recently received attention. However, the contribution of complement activation in CHIKV-induced pathologies has not been fully elucidated. The present study was undertaken to delineate the possible contribution of complement activation in CHIKV-induced disease progression. In this study, using plasma specimens of chikungunya patients in the acute, chronic, and recovered phases of infection, we explicated the involvement of complement activation in CHIKV disease progression by ELISAs and Bio-Plex assays. Correlation analysis was carried out to demonstrate interrelation among C1q-binding IgG-containing circulating immune complexes (CIC-C1q), complement activation fragments (C3a, C5a, sC5b-9), and complement-modulated pro-inflammatory cytokines (IL-1β, IL-18, IL-6, and TNF-α). We detected elevated complement activation fragments, CIC-C1q, and complement-modulated cytokines in the varied patient groups compared with the healthy controls, indicating persistent activation of the complement system. Furthermore, we observed statistically significant correlations among CIC-C1q with complement activation fragments and C3a with complement modulatory cytokines IL-1β, IL-6, and IL-18 during the CHIKV disease progression. Taken together, the current data provide insight into the plausible association between CICs, complement activation, subsequent complement modulatory cytokine expression, and CHIKV etiopathology.
Collapse
Affiliation(s)
- Rewati Kasbe
- Poliovirus Group (Former Avian Influenza), ICMR-National Institute of Virology, Pashan Campus, 130/1 Sus Road, Pashan, Pune, 411021, India
| | - Anuradha S Tripathy
- Dengue & Chikungunya Group, ICMR-National Institute of Virology, Pune, 411001, India
| | - Mohan R Wani
- National Centre for Cell Science, Pune, 411007, India
| | - Jayati Mullick
- Poliovirus Group (Former Avian Influenza), ICMR-National Institute of Virology, Pashan Campus, 130/1 Sus Road, Pashan, Pune, 411021, India.
| |
Collapse
|
3
|
Ghebrehiwet B, Joseph K, Kaplan AP. The bradykinin-forming cascade in anaphylaxis and ACE-inhibitor induced angioedema/airway obstruction. FRONTIERS IN ALLERGY 2024; 5:1302605. [PMID: 38332896 PMCID: PMC10850323 DOI: 10.3389/falgy.2024.1302605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Anaphylaxis is a potentially life-threatening multi-system allergic reaction to a biological trigger resulting in the release of potent inflammatory mediators from mast cells and basophils and causing symptoms in at least two organ systems that generally include skin, lungs, heart, or gastrointestinal tract in any combination. One exception is profound hypotension as an isolated symptom. There are two types of triggers of anaphylaxis: immunologic and non-Immunologic. Immunologic anaphylaxis is initiated when a foreign antigen directly binds to IgE expressed on mast cells or basophils and induces the release of histamine and other inflammatory substances resulting in vasodilation, vascular leakage, decreased peripheral vascular resistance, and heart muscle depression. If left untreated, death by shock (profound hypotension) or asphyxiation (airway obstruction) can occur. The non-immunologic pathway, on the other hand, can be initiated in many ways. A foreign substance can directly bind to receptors of mast cells and basophils leading to degranulation. There can be immune complex activation of the classical complement cascade with the release of anaphylatoxins C3a and C5a with subsequent recruitment of mast cells and basophils. Finally, hyperosmolar contrast agents can cause blood cell lysis, enzyme release, and complement activation, resulting in anaphylactoid (anaphylactic-like) symptoms. In this report we emphasize the recruitment of the bradykinin-forming cascade in mast cell dependent anaphylactic reactions as a potential mediator of severe hypotension, or airway compromise (asthma, laryngeal edema). We also consider airway obstruction due to inhibition of angiotensin converting enzyme with a diminished rate of endogenous bradykinin metabolism, leading not only to laryngeal edema, but massive tongue swelling with aspiration of secretions.
Collapse
Affiliation(s)
- Berhane Ghebrehiwet
- Division of Rheumatology, Allergy, and Clinical Immunology, SUNY-Stony Brook, Stony Brook, NY, United States
| | | | - Allen P. Kaplan
- Division of Pulmonary and Critical Care Medicine, The Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
4
|
Shughoury A, Sevgi DD, Ciulla TA. The complement system: a novel therapeutic target for age-related macular degeneration. Expert Opin Pharmacother 2023; 24:1887-1899. [PMID: 37691588 DOI: 10.1080/14656566.2023.2257604] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION With the recent FDA approvals of pegcetacoplan (SYFOVRE, Apellis Pharmaceuticals) and avacincaptad pegol (IZERVAY, Astellas Pharmaceuticals), modulation of the complement system has emerged as a promising therapeutic approach for slowing progression of geographic atrophy (GA) in AMD. AREAS COVERED This article reviews the current understanding of the complement system, its role in AMD, and the various complement-targeting therapies in development for the treatment of GA, including monoclonal antibodies, aptamers, protein analogs, and gene therapies. Approved and investigational agents have largely focused on interfering with the activity of complement components 3 and 5, owing to their central roles in the classical, lectin, and alternative complement pathways. Other investigational therapies have targeted formation of membrane attack complex (a terminal step in the complement cascade which leads to cell lysis), complement factors H and I (which serve regulatory functions in the alternative pathway), complement factors B and D (within the alternative pathway), and complement component 1 (within the classical pathway). Clinical trials investigating these agents are summarized, and the potential benefits and limitations of these therapies are discussed. EXPERT OPINION Targeting the complement system is a promising therapeutic approach for slowing the progression of GA in AMD, potentially improving visual outcomes. However, increased risk of exudative conversion must be considered, and further research is required to identify clinical criteria and best practices for initiating complement inhibitor therapy for GA.
Collapse
Affiliation(s)
- Aumer Shughoury
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Duriye D Sevgi
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Thomas A Ciulla
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
- Clearside Biomedical, Inc, Alpharetta, GA, USA
- Midwest Eye Institute, Carmel, IN, USA
| |
Collapse
|
5
|
Diamos AG, Pardhe MD, Bergeman MH, Kamzina AS, DiPalma MP, Aman S, Chaves A, Lowe K, Kilbourne J, Hogue IB, Mason HS. A self-binding immune complex vaccine elicits strong neutralizing responses against herpes simplex virus in mice. Front Immunol 2023; 14:1085911. [PMID: 37205110 PMCID: PMC10186352 DOI: 10.3389/fimmu.2023.1085911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/18/2023] [Indexed: 05/21/2023] Open
Abstract
Introduction It has been known for over half a century that mixing an antigen with its cognate antibody in an immune complex (IC) can enhance antigen immunogenicity. However, many ICs produce inconsistent immune responses, and the use of ICs in the development new vaccines has been limited despite the otherwise widespread success of antibody-based therapeutics. To address this problem, we designed a self-binding recombinant immune complex (RIC) vaccine which mimics the larger ICs generated during natural infection. Materials and methods In this study, we created two novel vaccine candidates: 1) a traditional IC targeting herpes simplex virus 2 (HSV-2) by mixing glycoprotein D (gD) with a neutralizing antibody (gD-IC); and 2) an RIC consisting of gD fused to an immunoglobulin heavy chain and then tagged with its own binding site, allowing self-binding (gD-RIC). We characterized the complex size and immune receptor binding characteristics in vitro for each preparation. Then, the in vivo immunogenicity and virus neutralization of each vaccine were compared in mice. Results gD-RIC formed larger complexes which enhanced C1q receptor binding 25-fold compared to gD-IC. After immunization of mice, gD-RIC elicited up to 1,000-fold higher gD-specific antibody titers compared to traditional IC, reaching endpoint titers of 1:500,000 after two doses without adjuvant. The RIC construct also elicited stronger virus-specific neutralization against HSV-2, as well as stronger cross-neutralization against HSV-1, although the proportion of neutralizing antibodies to total antibodies was somewhat reduced in the RIC group. Discussion This work demonstrates that the RIC system overcomes many of the pitfalls of traditional IC, providing potent immune responses against HSV-2 gD. Based on these findings, further improvements to the RIC system are discussed. RIC have now been shown to be capable of inducing potent immune responses to a variety of viral antigens, underscoring their broad potential as a vaccine platform.
Collapse
Affiliation(s)
- Andrew G. Diamos
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute at Arizona State University (ASU), School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | | | | | | | | | | | | | | | | | - Ian B. Hogue
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute at Arizona State University (ASU), School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Hugh S. Mason
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute at Arizona State University (ASU), School of Life Sciences, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
6
|
Ghosh M, Rana S. The anaphylatoxin C5a: Structure, function, signaling, physiology, disease, and therapeutics. Int Immunopharmacol 2023; 118:110081. [PMID: 36989901 DOI: 10.1016/j.intimp.2023.110081] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/06/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
The complement system is one of the oldest known tightly regulated host defense systems evolved for efficiently functioning cell-based immune systems and antibodies. Essentially, the complement system acts as a pivot between the innate and adaptive arms of the immune system. The complement system collectively represents a cocktail of ∼50 cell-bound/soluble glycoproteins directly involved in controlling infection and inflammation. Activation of the complement cascade generates complement fragments like C3a, C4a, and C5a as anaphylatoxins. C5a is the most potent proinflammatory anaphylatoxin, which is involved in inflammatory signaling in a myriad of tissues. This review provides a comprehensive overview of human C5a in the context of its structure and signaling under several pathophysiological conditions, including the current and future therapeutic applications targeting C5a.
Collapse
Affiliation(s)
- Manaswini Ghosh
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha 752050, India
| | - Soumendra Rana
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha 752050, India.
| |
Collapse
|
7
|
Sahu BS, Nguyen ME, Rodriguez P, Pallais JP, Ghosh V, Razzoli M, Sham YY, Salton SR, Bartolomucci A. The molecular identity of the TLQP-21 peptide receptor. Cell Mol Life Sci 2021; 78:7133-7144. [PMID: 34626205 PMCID: PMC8629782 DOI: 10.1007/s00018-021-03944-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 11/30/2022]
Abstract
The TLQP-21 neuropeptide has been implicated in functions as diverse as lipolysis, neurodegeneration and metabolism, thus suggesting an important role in several human diseases. Three binding targets have been proposed for TLQP-21: C3aR1, gC1qR and HSPA8. The aim of this review is to critically evaluate the molecular identity of the TLQP-21 receptor and the proposed multi-receptor mechanism of action. Several studies confirm a critical role for C3aR1 in TLQP-21 biological activity and a largely conserved mode of binding, receptor activation and signaling with C3a, its first-identified endogenous ligand. Conversely, data supporting a role of gC1qR and HSPA8 in TLQP-21 activity remain limited, with no signal transduction pathways being described. Overall, C3aR1 is the only receptor for which a necessary and sufficient role in TLQP-21 activity has been confirmed thus far. This conclusion calls into question the validity of a multi-receptor mechanism of action for TLQP-21 and should inform future studies.
Collapse
Affiliation(s)
- Bhavani S Sahu
- National Brain Research Centre, NH-8, Manesar, Gurugram, Haryana, 122052, India
| | - Megin E Nguyen
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA
- Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, USA
| | - Pedro Rodriguez
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA
| | - Jean Pierre Pallais
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA
| | - Vinayak Ghosh
- National Brain Research Centre, NH-8, Manesar, Gurugram, Haryana, 122052, India
| | - Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA
| | - Yuk Y Sham
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA
- Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, USA
| | - Stephen R Salton
- Departments of Neuroscience and Geriatrics and Palliative Medicine, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
8
|
Vivanco Gonzalez N, Oliveria JP, Tebaykin D, Ivison GT, Mukai K, Tsai MM, Borges L, Nadeau KC, Galli SJ, Tsai AG, Bendall SC. Mass Cytometry Phenotyping of Human Granulocytes Reveals Novel Basophil Functional Heterogeneity. iScience 2020; 23:101724. [PMID: 33205028 PMCID: PMC7653073 DOI: 10.1016/j.isci.2020.101724] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 08/18/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
Basophils, the rarest granulocyte, play critical roles in parasite- and allergen-induced inflammation. We applied mass cytometry (CyTOF) to simultaneously asses 44 proteins to phenotype and functionally characterize neutrophils, eosinophils, and basophils from 19 healthy donors. There was minimal heterogeneity seen in eosinophils and neutrophils, but data-driven analyses revealed four unique subpopulations within phenotypically basophilic granulocytes (PBG; CD45+HLA-DR-CD123+). Through CyTOF and fluorescence-activated cell sorting (FACS), we classified these four PBG subpopulations as (I) CD16lowFcεRIhighCD244high (88.5 ± 1.2%), (II) CD16highFcεRIhighCD244high (9.1 ± 0.4%), (III) CD16lowFcεRIlowCD244low (2.3 ± 1.3), and (IV) CD16highFcεRIlowCD244low (0.4 ± 0.1%). Prospective isolation confirmed basophilic-morphology of PBG I-III, but neutrophilic-morphology of PBG IV. Functional interrogation via IgE-crosslinking or IL-3 stimulation demonstrated that PBG I-II had significant increases in CD203c expression, whereas PBG III-IV remained unchanged compared with media-alone conditions. Thus, PBG III-IV could serve roles in non-IgE-mediated immunity. Our findings offer new perspectives in human basophil heterogeneity and the varying functional potential of these new subsets in health and disease.
Collapse
Affiliation(s)
- Nora Vivanco Gonzalez
- Department of Pathology, School of Medicine, Stanford University, Stanford Blood Center, 3373 Hillview Avenue Room 230A, Palo Alto, CA 94305, USA
| | - John-Paul Oliveria
- Department of Pathology, School of Medicine, Stanford University, Stanford Blood Center, 3373 Hillview Avenue Room 230A, Palo Alto, CA 94305, USA
- Department of Medicine, Division of Respirology, McMaster University, Hamilton, ON, L8S4K1, Canada
| | - Dmitry Tebaykin
- Department of Pathology, School of Medicine, Stanford University, Stanford Blood Center, 3373 Hillview Avenue Room 230A, Palo Alto, CA 94305, USA
| | - Geoffrey T. Ivison
- Department of Pathology, School of Medicine, Stanford University, Stanford Blood Center, 3373 Hillview Avenue Room 230A, Palo Alto, CA 94305, USA
| | - Kaori Mukai
- Department of Pathology, School of Medicine, Stanford University, Stanford Blood Center, 3373 Hillview Avenue Room 230A, Palo Alto, CA 94305, USA
- Sean N. Parker Center for Allergy Research, School of Medicine, Stanford University, Palo Alto, CA 94305, USA
| | - Mindy M. Tsai
- Department of Pathology, School of Medicine, Stanford University, Stanford Blood Center, 3373 Hillview Avenue Room 230A, Palo Alto, CA 94305, USA
- Sean N. Parker Center for Allergy Research, School of Medicine, Stanford University, Palo Alto, CA 94305, USA
| | - Luciene Borges
- Department of Pathology, School of Medicine, Stanford University, Stanford Blood Center, 3373 Hillview Avenue Room 230A, Palo Alto, CA 94305, USA
| | - Kari C. Nadeau
- Sean N. Parker Center for Allergy Research, School of Medicine, Stanford University, Palo Alto, CA 94305, USA
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, School of Medicine, Stanford University, Palo Alto, CA 94305, USA
| | - Stephen J. Galli
- Department of Pathology, School of Medicine, Stanford University, Stanford Blood Center, 3373 Hillview Avenue Room 230A, Palo Alto, CA 94305, USA
- Sean N. Parker Center for Allergy Research, School of Medicine, Stanford University, Palo Alto, CA 94305, USA
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Palo Alto, CA 94305, USA
| | - Albert G. Tsai
- Department of Pathology, School of Medicine, Stanford University, Stanford Blood Center, 3373 Hillview Avenue Room 230A, Palo Alto, CA 94305, USA
| | - Sean C. Bendall
- Department of Pathology, School of Medicine, Stanford University, Stanford Blood Center, 3373 Hillview Avenue Room 230A, Palo Alto, CA 94305, USA
| |
Collapse
|
9
|
Rowley JA, Reid RC, Poon EKY, Wu KC, Lim J, Lohman RJ, Hamidon JK, Yau MK, Halili MA, Durek T, Iyer A, Fairlie DP. Potent Thiophene Antagonists of Human Complement C3a Receptor with Anti-Inflammatory Activity. J Med Chem 2020; 63:529-541. [PMID: 31910011 DOI: 10.1021/acs.jmedchem.9b00927] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Structure-activity relationships for a series of small-molecule thiophenes resulted in potent and selective antagonism of human Complement C3a receptor. The compounds are about 100-fold more potent than the most reported antagonist SB290157. A new compound JR14a was among the most potent of the new antagonists in vitro, assessed by (a) inhibition of intracellular calcium release (IC50 10 nM) induced in human monocyte-derived macrophages by 100 nM C3a, (b) inhibition of β-hexosaminidase secretion (IC50 8 nM) from human LAD2 mast cells degranulated by 100 nM C3a, and (c) selectivity for human C3aR over C5aR. JR14a was metabolically stable in rat plasma and in rat liver microsomes and efficacious in rats when given orally to suppress rat paw inflammation, macrophage and mast cell activation, and histopathology induced by intraplantar paw administration of a C3aR agonist. Potent C3aR antagonists are now available for interrogating C3a receptor activation and suppressing C3aR-mediated inflammation in mammalian physiology and disease.
Collapse
|
10
|
Sahu BS, Rodriguez P, Nguyen ME, Han R, Cero C, Razzoli M, Piaggi P, Laskowski LJ, Pavlicev M, Muglia L, Mahata SK, O'Grady S, McCorvy JD, Baier LJ, Sham YY, Bartolomucci A. Peptide/Receptor Co-evolution Explains the Lipolytic Function of the Neuropeptide TLQP-21. Cell Rep 2019; 28:2567-2580.e6. [PMID: 31484069 PMCID: PMC6753381 DOI: 10.1016/j.celrep.2019.07.101] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 06/11/2019] [Accepted: 07/26/2019] [Indexed: 12/24/2022] Open
Abstract
Structural and functional diversity of peptides and GPCR result from long evolutionary processes. Even small changes in sequence can alter receptor activation, affecting therapeutic efficacy. We conducted a structure-function relationship study on the neuropeptide TLQP-21, a promising target for obesity, and its complement 3a receptor (C3aR1). After having characterized the TLQP-21/C3aR1 lipolytic mechanism, a homology modeling and molecular dynamics simulation identified the TLQP-21 binding motif and C3aR1 binding site for the human (h) and mouse (m) molecules. mTLQP-21 showed enhanced binding affinity and potency for hC3aR1 compared with hTLQP-21. Consistently, mTLQP-21, but not hTLQP-21, potentiates lipolysis in human adipocytes. These findings led us to uncover five mutations in the C3aR1 binding pocket of the rodent Murinae subfamily that are causal for enhanced calculated affinity and measured potency of TLQP-21. Identifying functionally relevant peptide/receptor co-evolution mechanisms can facilitate the development of innovative pharmacotherapies for obesity and other diseases implicating GPCRs.
Collapse
Affiliation(s)
- Bhavani S Sahu
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6(th) St. SE, Minneapolis, MN, USA
| | - Pedro Rodriguez
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6(th) St. SE, Minneapolis, MN, USA
| | - Megin E Nguyen
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6(th) St. SE, Minneapolis, MN, USA
| | - Ruijun Han
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6(th) St. SE, Minneapolis, MN, USA
| | - Cheryl Cero
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6(th) St. SE, Minneapolis, MN, USA
| | - Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6(th) St. SE, Minneapolis, MN, USA
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes, Digestive and Kidney Diseases, NIH, Phoenix, AZ, USA
| | - Lauren J Laskowski
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mihaela Pavlicev
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Louis Muglia
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sushil K Mahata
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Scott O'Grady
- Department of Animal Science, University of Minnesota, 480 Haecker Hall, 1364 Eckles Avenue, St. Paul, MN, USA
| | - John D McCorvy
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Leslie J Baier
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes, Digestive and Kidney Diseases, NIH, Phoenix, AZ, USA
| | - Yuk Y Sham
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6(th) St. SE, Minneapolis, MN, USA; Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, MN, USA
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota, 2231 6(th) St. SE, Minneapolis, MN, USA.
| |
Collapse
|
11
|
Frazier KS, Obert LA. Drug-induced Glomerulonephritis: The Spectre of Biotherapeutic and Antisense Oligonucleotide Immune Activation in the Kidney. Toxicol Pathol 2018; 46:904-917. [PMID: 30089413 DOI: 10.1177/0192623318789399] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prevalence of immune-mediated glomerulonephritis has increased in preclinical toxicity studies, with more frequent use of biotherapeutic agents (especially antigenic humanized molecules) and antisense oligonucleotide (ASO) therapies. Immune complex disease affects a small number of study monkeys, often correlates with antidrug antibody (ADA) titers, and occurs at a dose that favors immune complex formation or impedes clearance. While preclinical glomerulonephritis often fails to correlate with evidence of glomerular or vascular injury in human clinical trials and is not considered predictive, additional animal investigative immunohistochemical work may be performed to substantiate evidence for immune complex pathogenesis. While ADA is most commonly encountered as a predisposing factor with biotherapeutic agents, complement activation may occur without circulating complexes, and other mechanisms of non-ADA immune-mediated glomerulonephritis have been observed including nonendogenous immune aggregates and immunoregulatory pharmacology. Although glomerulonephritis associated with oligonucleotide therapies has been noted occasionally in preclinical studies and more rarely with human patients, pathophysiologic mechanisms involved appear to be different between species and preclinical cases are not considered predictive for humans. ADA is not involved in oligonucleotide-associated cases, and complement fixation plays a more important role in monkeys. Recent screening of ASOs for proinflammatory activity appears to have decreased glomerulonephritis incidence preclinically.
Collapse
|
12
|
Basu A, Hong J, Ferraz N. Hemocompatibility of Ca2+
-Crosslinked Nanocellulose Hydrogels: Toward Efficient Management of Hemostasis. Macromol Biosci 2017; 17. [DOI: 10.1002/mabi.201700236] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/10/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Alex Basu
- Nanotechnology and Functional Materials; Department of Engineering Sciences; Uppsala University; Box 534 75121 Uppsala Sweden
| | - Jaan Hong
- Department of Immunology; Genetics and Pathology; Rudbeck Laboratory C5; Uppsala University; 75185 Uppsala Sweden
| | - Natalia Ferraz
- Nanotechnology and Functional Materials; Department of Engineering Sciences; Uppsala University; Box 534 75121 Uppsala Sweden
| |
Collapse
|
13
|
Huang X, Wang R, Lu T, Zhou D, Zhao W, Sun S, Zhao C. Heparin-Like Chitosan Hydrogels with Tunable Swelling Behavior, Prolonged Clotting Times, and Prevented Contact Activation and Complement Activation. Biomacromolecules 2016; 17:4011-4020. [DOI: 10.1021/acs.biomac.6b01386] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Xuelian Huang
- College of Polymer Science
and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Rui Wang
- College of Polymer Science
and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Ting Lu
- College of Polymer Science
and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Dongxu Zhou
- College of Polymer Science
and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Weifeng Zhao
- College of Polymer Science
and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Shudong Sun
- College of Polymer Science
and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Changsheng Zhao
- College of Polymer Science
and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
14
|
Wouters D, Zeerleder S. Complement inhibitors to treat IgM-mediated autoimmune hemolysis. Haematologica 2016; 100:1388-95. [PMID: 26521297 DOI: 10.3324/haematol.2015.128538] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Complement activation in autoimmune hemolytic anemia may exacerbate extravascular hemolysis and may occasionally result in intravascular hemolysis. IgM autoantibodies as characteristically found in cold autoantibody autoimmune hemolytic anemia, in cold agglutinin disease but also in a considerable percentage of patients with warm autoantibodies are very likely to activate complement in vivo. Therapy of IgM-mediated autoimmune hemolytic anemia mainly aims to decrease autoantibody production. However, most of these treatments require time to become effective and will not stop immediate ongoing complement-mediated hemolysis nor prevent hemolysis of transfused red blood cells. Therefore pharmacological inhibition of the complement system might be a suitable approach to halt or at least attenuate ongoing hemolysis and improve the recovery of red blood cell transfusion in autoimmune hemolytic anemia. In recent years, several complement inhibitors have become available in the clinic, some of them with proven efficacy in autoimmune hemolytic anemia. In the present review, we give a short introduction on the pathogenesis of autoimmune hemolytic anemia, followed by an overview on the complement system with a special focus on its regulation. Finally, we will discuss complement inhibitors with regard to their potential efficacy to halt or attenuate hemolysis in complement-mediated autoimmune hemolytic anemia.
Collapse
Affiliation(s)
- Diana Wouters
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, the Netherlands
| | - Sacha Zeerleder
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, the Netherlands Department of Hematology, Academic Medical Center, University of Amsterdam, the Netherlands
| |
Collapse
|
15
|
Mahajan SD, Tutino VM, Redae Y, Meng H, Siddiqui A, Woodruff TM, Jarvis JN, Hennon T, Schwartz S, Quigg RJ, Alexander JJ. C5a induces caspase-dependent apoptosis in brain vascular endothelial cells in experimental lupus. Immunology 2016; 148:407-19. [PMID: 27213693 DOI: 10.1111/imm.12619] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/06/2016] [Accepted: 05/10/2016] [Indexed: 12/18/2022] Open
Abstract
Blood-brain barrier (BBB) dysfunction complicates central nervous system lupus, an important aspect of systemic lupus erythematosus. To gain insight into the underlying mechanism, vascular corrosion casts of brain were generated from the lupus mouse model, MRL/lpr mice and the MRL/MpJ congenic controls. Scanning electron microscopy of the casts showed loss of vascular endothelial cells in lupus mice compared with controls. Immunostaining revealed a significant increase in caspase 3 expression in the brain vascular endothelial cells, which suggests that apoptosis could be an important mechanism causing cell loss, and thereby loss of BBB integrity. Complement activation occurs in lupus resulting in increased generation of circulating C5a, which caused the endothelial layer to become 'leaky'. In this study, we show that C5a and lupus serum induced apoptosis in cultured human brain microvascular endothelial cells (HBMVECs), whereas selective C5a receptor 1 (C5aR1) antagonist reduced apoptosis in these cells, demonstrating C5a/C5aR1-dependence. Gene expression of initiator caspases, caspase 1 and caspase 8, and pro-apoptotic proteins death-associated protein kinase 1, Fas-associated protein (FADD), cell death-inducing DNA fragmentation factor 45 000 MW subunit A-like effector B (CIDEB) and BCL2-associated X protein were increased in HBMVECs treated with lupus serum or C5a, indicating that both the intrinsic and extrinsic apoptotic pathways could be critical mediators of brain endothelial cell apoptosis in this setting. Overall, our findings suggest that C5a/C5aR1 signalling induces apoptosis through activation of FADD, caspase 8/3 and CIDEB in brain endothelial cells in lupus. Further elucidation of the underlying apoptotic mechanisms mediating the reduced endothelial cell number is important in establishing the potential therapeutic effectiveness of C5aR1 inhibition that could prevent and/or reduce BBB alterations and preserve the physiological function of BBB in central nervous system lupus.
Collapse
Affiliation(s)
| | - Vincent M Tutino
- Toshiba Vascular Stroke Center, Biomedical Engineering Department, University at Buffalo, Buffalo, NY, USA
| | - Yonas Redae
- Department of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Hui Meng
- Toshiba Vascular Stroke Center, Biomedical Engineering Department, University at Buffalo, Buffalo, NY, USA
| | - Adnan Siddiqui
- Toshiba Vascular Stroke Center, Biomedical Engineering Department, University at Buffalo, Buffalo, NY, USA
| | - Trent M Woodruff
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD, Australia
| | - James N Jarvis
- Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| | - Teresa Hennon
- Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| | | | - Richard J Quigg
- Department of Medicine, University at Buffalo, Buffalo, NY, USA
| | | |
Collapse
|
16
|
van Oeveren W, Wildevuur C. Review article : Blood compatibility of cardiopulmonary bypass circuits. Perfusion 2016. [DOI: 10.1177/026765918700200402] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
17
|
Aye KS, Charngkaew K, Win N, Wai KZ, Moe K, Punyadee N, Thiemmeca S, Suttitheptumrong A, Sukpanichnant S, Prida M, Halstead SB. Pathologic highlights of dengue hemorrhagic fever in 13 autopsy cases from Myanmar. Hum Pathol 2014; 45:1221-33. [PMID: 24767772 DOI: 10.1016/j.humpath.2014.01.022] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 01/20/2014] [Accepted: 01/30/2014] [Indexed: 12/31/2022]
Abstract
Vascular permeability, thrombocytopenia, liver pathology, complement activation, and altered hemostasis accompanying a febrile disease are the hallmarks of the dengue hemorrhagic fever/dengue shock syndrome, a major arthropod-borne viral disease that causes significant morbidity and mortality throughout tropical countries. We studied tissues from 13 children who died of acute dengue hemorrhagic fever/dengue shock syndrome at the Childrens' Hospital, Yangon, Myanmar. Dengue viral RNA from each of the 4 dengue viruses (DENVs) was detected by reverse transcriptase polymerase chain reaction in 11 cases, and dengue viral proteins (envelope, NS1, or NS3) were detected in 1 or more tissues from all 13 cases. Formalin-fixed and frozen tissues were studied for evidence of virus infection using monoclonal antibodies against DENV structural and nonstructural antigens (E, NS1, and nonsecreting NS3). In the liver, DENV infection occurred in hepatocytes and Kupffer cells but not in endothelial cells. Liver damage was associated with deposition on hepatocytes of complement components of both classical and alternative pathways. Evidence of dengue viral replication was observed in macrophage-like cells in spleens and lymph nodes. No dengue antigens were detected in endothelial cells in any organ. Germinal centers of the spleen and lymph nodes showed a marked reduction in the number of lymphocytes that were replaced by eosinophilic deposits, which contained dengue antigens as well as immunoglobulins, and complement components (C3, C1q, and C9). The latter findings had previously been reported but overlooked as a diagnostic feature.
Collapse
Affiliation(s)
- Khin Saw Aye
- Department of Medical Research (Lower Myanmar), Yangon, Myanmar
| | - Komgrid Charngkaew
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ne Win
- National Health Laboratory, Yangon, Myanmar
| | - Kyaw Zin Wai
- Intensive Care Unit, Yangon Children Hospital, Yangon, Myanmar
| | - Kyaw Moe
- Department of Medical Research (Lower Myanmar), Yangon, Myanmar
| | - Nuntaya Punyadee
- Dengue Hemorrhagic Fever Research Unit, Mahidol University, Bangkok, Thailand
| | - Somchai Thiemmeca
- Dengue Hemorrhagic Fever Research Unit, Mahidol University, Bangkok, Thailand
| | - Aroonroong Suttitheptumrong
- Division of Molecular Medicine, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sanya Sukpanichnant
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Malasit Prida
- Dengue Hemorrhagic Fever Research Unit, Mahidol University, Bangkok, Thailand; Medical Biotechnology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok, Thailand.
| | - Scott B Halstead
- Dengue Vaccine Initiative, International Vaccine Institute, Seoul, South Korea.
| |
Collapse
|
18
|
Klos A, Wende E, Wareham KJ, Monk PN. International Union of Basic and Clinical Pharmacology. [corrected]. LXXXVII. Complement peptide C5a, C4a, and C3a receptors. Pharmacol Rev 2013; 65:500-43. [PMID: 23383423 DOI: 10.1124/pr.111.005223] [Citation(s) in RCA: 189] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The activation of the complement cascade, a cornerstone of the innate immune response, produces a number of small (74-77 amino acid) fragments, originally termed anaphylatoxins, that are potent chemoattractants and secretagogues that act on a wide variety of cell types. These fragments, C5a, C4a, and C3a, participate at all levels of the immune response and are also involved in other processes such as neural development and organ regeneration. Their primary function, however, is in inflammation, so they are important targets for the development of antiinflammatory therapies. Only three receptors for complement peptides have been found, but there are no satisfactory antagonists as yet, despite intensive investigation. In humans, there is a single receptor for C3a (C3a receptor), no known receptor for C4a, and two receptors for C5a (C5a₁ receptor and C5a₂ receptor). The most recently characterized receptor, the C5a₂ receptor (previously known as C5L2 or GPR77), has been regarded as a passive binding protein, but signaling activities are now ascribed to it, so we propose that it be formally identified as a receptor and be given a name to reflect this. Here, we describe the complex biology of the complement peptides, introduce a new suggested nomenclature, and review our current knowledge of receptor pharmacology.
Collapse
Affiliation(s)
- Andreas Klos
- Department for Medical Microbiology, Medical School Hannover, Hannover, Germany
| | | | | | | |
Collapse
|
19
|
Ikeda K, Fukuda N, Ueno T, Endo M, Kobayashi N, Soma M, Matsumoto K. Role of complement 3a in the growth of mesangial cells from stroke-prone spontaneously hypertensive rats. Clin Exp Hypertens 2013; 36:58-63. [PMID: 23713944 DOI: 10.3109/10641963.2013.789042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Vascular smooth muscle cells (VSMCs) derived from spontaneously hypertensive rats (SHR) show exaggerated growth with a synthetic phenotype and angiotensin II (Ang II) production associated with increased production of complement (C3). We hypothesized that C3 is involved in the growth of mesangial cells (MCs) from hypertensive rats. We examined the effects of a C3a receptor inhibitor on proliferation, phenotype and Ang II generation in MCs from stroke prone-spontaneously hypertensive rats (SHR)-SP, SHR and Wistar-Kyoto (WKY) rats. Expression of C3 and C3a receptor were evaluated by immunohistochemical staining of the renal cortex. We examined the effects of the C3a inhibitor, SB290157, on proliferation, the expression of phenotype-marker mRNAs and Ang II production in cells from SHR-SP, SHR and WKY rats. Immunostaining of C3 was stronger in SHR and SHRSP glomeruli. MCs from SHR-SP and SHR abundantly express pre-pro C3 mRNA. SB290157 significantly inhibited basal DNA synthesis and proliferation of MCs from SHR-SP and SHR. Expression of osteopontin mRNA in MCs from SHR-SP and SHR was decreased with SB290157 treatment, whereas MC basal expression of α-SMA mRNA was decreased. SB290157 significantly decreased the production of Ang II in MCs from SHR-SP and SHR. Endogenous C3a promotes exaggerated growth with a synthetic phenotype and the production of Ang II in MCs from SHR-SP and SHR. The C3 and C3a receptor system may primarily be involved in the pathogenesis of renal remodeling in hypertensive rats.
Collapse
Affiliation(s)
- Kazuya Ikeda
- Division of Nephrology, Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine , Tokyo , Japan
| | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Maternal recognition of fetal antigens is well-documented. Antibodies directed against fetal antigens are the rule rather than the exception. Maternal antibodies seem to bind fetal antigens at the placental level and apparently activate the complement system even in normal pregnancy, yet multiple studies confirm an increase in C1q, C4, C3 and CH50 levels in pregnancy and an absence of complement activation. More sensitive assays and a broadened concept of the timing of crucial immunologic events may lead to a greater understanding of the importance of the complement system in pregnancy.
Collapse
|
21
|
Sanderson SD, Thoman ML, Kis K, Virts EL, Herrera EB, Widmann S, Sepulveda H, Phillips JA. Innate immune induction and influenza protection elicited by a response-selective agonist of human C5a. PLoS One 2012; 7:e40303. [PMID: 22792270 PMCID: PMC3391237 DOI: 10.1371/journal.pone.0040303] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 06/04/2012] [Indexed: 12/20/2022] Open
Abstract
The anaphylatoxin C5a is an especially potent mediator of both local and systemic inflammation. However, C5a also plays an essential role in mucosal host defense against bacterial, viral, and fungal infection. We have developed a response-selective agonist of human C5a, termed EP67, which retains the immunoenhancing activity of C5a at the expense of its inflammatory, anaphylagenic properties. EP67 insufflation results in the rapid induction of pulmonary cytokines and chemokines. This is followed by an influx of innate immune effector cells, including neutrophils, NK cells, and dendritic cells. EP67 exhibits both prophylactic and therapeutic protection when tested in a murine model of influenza A infection. Mice treated with EP67 within a twenty-four hour window of non-lethal infection were significantly protected from influenza-induced weight loss. Furthermore, EP67 delivered twenty-four hours after lethal infection completely blocked influenza-induced mortality (0% vs. 100% survival). Since protection based on innate immune induction is not restricted to any specific pathogen, EP67 may well prove equally efficacious against a wide variety of possible viral, bacterial, and fungal pathogens. Such a strategy could be used to stop the worldwide spread of emergent respiratory diseases, including but not limited to novel strains of influenza.
Collapse
Affiliation(s)
- Sam D. Sanderson
- School of Allied Health Professions, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Marilyn L. Thoman
- Sidney Kimmel Cancer Center, San Diego, California, United States of America
- Biosciences Center, San Diego State University, San Diego, California, United States of America
| | - Kornelia Kis
- Biosciences Center, San Diego State University, San Diego, California, United States of America
| | - Elizabeth L. Virts
- Sidney Kimmel Cancer Center, San Diego, California, United States of America
- Biosciences Center, San Diego State University, San Diego, California, United States of America
| | - Edgar B. Herrera
- Biosciences Center, San Diego State University, San Diego, California, United States of America
| | | | | | - Joy A. Phillips
- Sidney Kimmel Cancer Center, San Diego, California, United States of America
- Biosciences Center, San Diego State University, San Diego, California, United States of America
| |
Collapse
|
22
|
Role of complement 3a in the synthetic phenotype and angiotensin II-production in vascular smooth muscle cells from spontaneously hypertensive rats. Am J Hypertens 2012; 25:284-9. [PMID: 22089112 PMCID: PMC3280391 DOI: 10.1038/ajh.2011.214] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background Spontaneously hypertensive rats (SHR)-derived vascular smooth muscle cells (VSMCs) show exaggerated growth with a synthetic phenotype and angiotensin II (Ang II)-production. To evaluate the contribution of complement 3 (C3) or C3a toward these abnormalities in SHR, we examined effects of a C3a receptor inhibitor on proliferation, phenotype, and Ang II-production in VSMCs from SHR and Wistar–Kyoto (WKY) rats. Methods Expression of pre-pro-C3 messenger RNA (mRNA) and C3 protein was evaluated by reverse transcription-PCR and western blot analyses, and C3a receptor mRNA was evaluated by reverse transcription-PCR analysis in quiescent VSMCs from SHR and WKY rats. We examined the effects of the C3a inhibitor, SB290157, on proliferation and the expression of phenotype-marker and Krueppel-like factor 5 (KLF-5) mRNAs in VSMCs from SHR and WKY rats. We examined effects of C3a receptor inhibitor, SB290157, on Ang II-production in conditioned medium of VSMCs from SHR and WKY rats by a radioimmunoassay. Results Expression of pre-pro-C3 mRNA and C3 protein was significantly higher in SHR VSMCs than WKY VSMCs. SB290157 significantly inhibited proliferation of VSMCs from SHR, but not in cells from WKY rats. Relative to WKY VSMCs, SB290157 significantly increased the low expression of SM22α mRNA and decreased the high expression of osteopontin mRNA in SHR VSMCs. SB290157 significantly decreased the high expression of KLF-5 and Ang II-production in VSMCs from SHR, but not in cells from WKY rats. Conclusions C3a induces exaggerated growth, a synthetic phenotype and Ang II-production in SHR-derived VSMCs. C3a may be primarily involved in cardiovascular remodeling in hypertension.
Collapse
|
23
|
Woodruff RS, Sullenger B, Becker RC. The many faces of the contact pathway and their role in thrombosis. J Thromb Thrombolysis 2011; 32:9-20. [PMID: 21404067 DOI: 10.1007/s11239-011-0578-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Understanding inherent differences between thrombosis and hemostasis in the vascular system are critical to developing safe and effective anticoagulants. To this end, constituents of the contact activated and intrinsic pathway of coagulation appear to be involved in pathological thrombus formation, but are not required for normal hemostasis. In addition to coagulation, activation of the contact system is involved in fibrinolytic, inflammatory, and angiogenic processes that can also contribute to the thrombotic environment. This review discusses the role of the contact system in these processes, and highlights the potential of FXII and FXI as safer targets for antithrombotic therapy.
Collapse
Affiliation(s)
- Rebecca S Woodruff
- Duke Translational Research Institute, Duke University School of Medicine, Durham, NC, USA
| | | | | |
Collapse
|
24
|
Pathogenesis of implant-associated infection: the role of the host. Semin Immunopathol 2011; 33:295-306. [DOI: 10.1007/s00281-011-0275-7] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 02/14/2011] [Indexed: 01/30/2023]
|
25
|
Chimote G, Banerjee R. In vitro evaluation of inhalable isoniazid-loaded surfactant liposomes as an adjunct therapy in pulmonary tuberculosis. J Biomed Mater Res B Appl Biomater 2010; 94:1-10. [PMID: 20524179 DOI: 10.1002/jbm.b.31608] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
In this study, exogenous pulmonary surfactant was evaluated as an inhalable drug carrier for antitubercular drug isoniazid (INH). Isoniazid-entrapped liposomes of dipalmitoylphosphatidylcholine (DPPC) (the most abundant lipid of lung surfactant and exogenous surfactant) were developed and evaluated for size, drug entrapment, release, in vitro alveolar deposition, biocompatibility, antimycobacterial activity, and pulmonary surfactant action. Isoniazid-entrapped DPPC liposomes were about 750 nm in diameter and had entrapment efficiency of 36.7% +/- 1.8%. Sustained release of INH from DPPC liposomes was observed over 24 h. In vitro alveolar deposition efficiency using the twin impinger exhibited approximately 25-27% INH deposition in the alveolar chamber upon one minute nebulization using a jet nebulizer. At 37 degrees C, the formulation had better pulmonary surfactant function with quicker reduction of surface tension on adsorption (36.7 +/- 0.4 mN/m) than DPPC liposomes (44.7 +/- 0.6 mN/m) and 87% airway patency was exhibited by the formulation in a capillary surfactometer. The formulation was biocompatible and had antimycobacterial activity. The isoniazid-entrapped DPPC liposomes could fulfill the dual purpose of pulmonary drug delivery and alveolar stabilization due to antiatelectatic effect of the surfactant action which can improve the reach of antitubercular drug INH to the alveoli.
Collapse
Affiliation(s)
- G Chimote
- School of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400 076, India
| | | |
Collapse
|
26
|
Abstract
Previous studies using blocking antibodies suggested that bone marrow (BM)-derived C3 is required for efficient osteoclast (OC) differentiation, and that C3 receptors are involved in this process. However, the detailed underlying mechanism and the possible involvement of other complement receptors remain unclear. In this report, we found that C3(-/-) BM cells exhibited lower RANKL/OPG expression ratios, produced smaller amounts of macrophage colony-stimulating factor and interleukin-6 (IL-6), and generated significantly fewer OCs than wild-type (WT) BM cells. During differentiation, in addition to C3, WT BM cells locally produced all other complement components required to activate C3 and to generate C3a/C5a through the alter-native pathway, which is required for efficient OC differentiation. Abrogating C3aR/C5aR activity either genetically or pharmaceutically suppressed OC generation, while stimulating WT or C3(-/-) BM cells with exogenous C3a and/or C5a augmented OC differentiation. Furthermore, supplementation with IL-6 rescued OC generation from C3(-/-) BM cells, and neutralizing antibodies to IL-6 abolished the stimulatory effects of C3a/C5a on OC differentiation. These data indicate that during OC differentiation, BM cells locally produce components, which are activated through the alternative pathway to regulate OC differentiation. In addition to C3 receptors, C3aR/C5aR also regulate OC differentiation, at least in part, by modulating local IL-6 production.
Collapse
|
27
|
Scully CCG, Blakeney JS, Singh R, Hoang HN, Abbenante G, Reid RC, Fairlie DP. Selective Hexapeptide Agonists and Antagonists for Human Complement C3a Receptor. J Med Chem 2010; 53:4938-48. [DOI: 10.1021/jm1003705] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Conor C. G. Scully
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jade S. Blakeney
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Ranee Singh
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Huy N. Hoang
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Giovanni Abbenante
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Robert C. Reid
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - David P. Fairlie
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
28
|
Abstract
The complement system plays a crucial role in the innate defense against common pathogens. Activation of complement leads to robust and efficient proteolytic cascades, which terminate in opsonization and lysis of the pathogen as well as in the generation of the classical inflammatory response through the production of potent proinflammatory molecules. More recently, however, the role of complement in the immune response has been expanded due to observations that link complement activation to adaptive immune responses. It is now appreciated that complement is a functional bridge between innate and adaptive immune responses that allows an integrated host defense to pathogenic challenges. As such, a study of its functions allows insight into the molecular underpinnings of host-pathogen interactions as well as the organization and orchestration of the host immune response. This review attempts to summarize the roles that complement plays in both innate and adaptive immune responses and the consequences of these interactions on host defense.
Collapse
|
29
|
Clark CJ, Chettibi S, Young JD, Greer IA, Lyall F. Locomotion of Human Neutrophils in Response to Plasma and Serum of Women with Preeclampsia. Hypertens Pregnancy 2009. [DOI: 10.3109/10641959609015706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
30
|
Abstract
Perturbation of the serum complement system in glomerulonephritis can be the result of classical pathway activation by immune complexes or of C3 activation by the C3-C5 convertase stabilized by the C3 nephritic factor. Low C3 levels can also be the result of diminished C3 synthesis and possibly, in certain circumstances, of C3 convertases deposited on capillary walls. In glomerulonephritis the complement profile is helpful in diagnosis, in following the course of therapy and in providing insights into pathogenesis. Complement profiles must be interpreted recognizing that a pattern resembling classical pathway activation can be produced by idiopathic nephrotic syndrome, that hypogammaglobulinemia can reduce Clq levels, and that a primary deficiency of factor H or I or both, will secondarily produce subnormal levels of C3 and factor B. With these caveats, the complement profiles typical of systemic lupus erythematosus, membranoproliferative glomerulonephritis, acute glomerulonephritis, acquired Cl inhibitor deficiency, and hypocomplementemic vasculitis syndrome are described.
Collapse
|
31
|
Graham M, Shin DH, Smith SL. Molecular and expression analysis of complement component C5 in the nurse shark (Ginglymostoma cirratum) and its predicted functional role. FISH & SHELLFISH IMMUNOLOGY 2009; 27:40-9. [PMID: 19410004 PMCID: PMC2734960 DOI: 10.1016/j.fsi.2009.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 04/13/2009] [Accepted: 04/21/2009] [Indexed: 05/07/2023]
Abstract
We present the complete cDNA sequence of shark (Ginglymostoma cirratum) pro-C5 and its molecular characterization with a descriptive analysis of the structural elements necessary for its potential functional role as a potent mediator of inflammation (fragment C5a) and initiator molecule (fragment C5b) for the assembly of the membrane attack complex (MAC) upon activation by C5 convertase. In mammals the three complement activation cascades, the classical, alternative and lectin pathways, converge at the activation of C3, a pivotal complement protein. It is, however, the subsequent activation of the next complement component, C5, which is the focal point at which the initiation of the terminal lytic pathway takes place and involves the stepwise assembly of the MAC. The effector cytolytic function of complement occurs with the insertion of MAC into target membranes causing dough-nut like holes and cell leakage. The lytic activity of shark complement results in structurally similar holes in target membranes suggesting the assembly of a shark MAC that likely involves a functional analogue of C5. The composition of shark MAC remains unresolved and to date conclusive evidence has been lacking for shark C5. The gene has not been cloned nor has the serum protein been characterized for any elasmobranch species. This report is the first to confirm the presence of C5 homologue in the shark. GcC5 is remarkably similar to human C5 in overall structure and domain arrangement. The GcC5 cDNA measured 5160-bp with 5' and 3' UTRs of 35 bp and 79 bp, respectively. Structural analysis of the derived protein sequence predicts a molecule that is a two-chain structure which lacks a thiolester bond and contains a C5 convertase cleavage site indicating that activation will generate two peptides, akin to C5b and C5a. The putative GcC5 molecule also contains the C-terminal C345C/Netrin module that characterizes C3, C4 and C5. Multiple alignment of deduced amino acid sequences shows that GcC5 shares more amino acid identities/similarities with mammals than that with bony fish. We conclude that at the time of emergence of sharks the elaborate mosaic structure of C5 had already evolved.
Collapse
Affiliation(s)
- Matthew Graham
- Department of Biological Sciences, Florida International University, University Park, Miami, Fl, 33199, USA
| | - Dong-Ho Shin
- Department of Biological Sciences and Comparative Immunology Institute, Florida International University, Miami, Fl, 33199, USA
| | - Sylvia L. Smith
- Department of Biological Sciences and Comparative Immunology Institute, Florida International University, Miami, Fl, 33199, USA
| |
Collapse
|
32
|
Loa loa Microfilariae evade complement attack in vivo by acquiring regulatory proteins from host plasma. Infect Immun 2009; 77:3886-93. [PMID: 19528206 DOI: 10.1128/iai.01583-08] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Loa loa is a filarial nematode that infects humans. The adults live in subcutaneous tissues and produce microfilariae that live for several weeks in the blood circulation in order to be transmitted to another person via blood meals of a dipterian vector. As microfilariae live in continuous contact with plasma, it is obvious that they evade the complement system. We studied markers of complement activation and signs of complement regulation on Loa loa microfilariae in vivo. The microfilariae were isolated from anticoagulated blood samples of a Loa loa-infected Caucasian patient. C1q and some mannose-binding lectin but only a limited amount of C3b or C4b fragments and practically no C5 or C5b-9 were present on the microfilariae. The covalently microfilaria-bound C3 and C4 depositions were mainly inactive iC3b, C3c, and iC4b fragments indicating that microfilariae had regulated complement activation in vivo. Also, in vitro deposition of C3b onto the microfilariae upon serum exposure was limited. The patient-isolated microfilariae were found to carry the host complement regulators factor H and C4b-binding protein on the outermost layer, so called sheath. The microfilaria-bound factor H was functionally active. Binding of the complement regulators to the microfilariae was confirmed in vitro using (125)I-labeled factor H and C4b-binding protein. In conclusion, our study shows that Loa loa microfilariae block complement activation and acquire the host complement regulators factor H and C4b-binding protein in blood circulation. This is the first time that binding of complement regulators onto nonviral pathogens has been demonstrated to occur in humans in vivo.
Collapse
|
33
|
Mueller-Ortiz SL, Wang D, Morales JE, Li L, Chang JY, Wetsel RA. Targeted disruption of the gene encoding the murine small subunit of carboxypeptidase N (CPN1) causes susceptibility to C5a anaphylatoxin-mediated shock. THE JOURNAL OF IMMUNOLOGY 2009; 182:6533-9. [PMID: 19414808 DOI: 10.4049/jimmunol.0804207] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Carboxypeptidase N (CPN) is a plasma zinc metalloprotease, which consists of two enzymatically active small subunits (CPN1) and two large subunits (CPN2) that protect the protein from degradation. Historically, CPN has been implicated as a major regulator of inflammation by its enzymatic cleavage of functionally important arginine and lysine amino acids from potent phlogistic molecules, such as the complement anaphylatoxins C3a and C5a. Because of no known complete CPN deficiencies, the biological impact of CPN in vivo has been difficult to evaluate. Here, we report the generation of a mouse with complete CPN deficiency by targeted disruption of the CPN1 gene. CPN1(-/-) mice were hypersensitive to lethal anaphylactic shock due to acute complement activation by cobra venom factor. This hypersensitivity was completely resolved in CPN1(-/-)/C5aR(-/-) but not in CPN1(-/-)/C3aR(-/-) mice. Moreover, CPN1(-/-) mice given C5a i.v., but not C3a, experienced 100% mortality. This C5a-induced mortality was reduced to 20% when CPN1(-/-) mice were treated with an antihistamine before C5a challenge. These studies describe for the first time a complete deficiency of CPN and demonstrate 1) that CPN plays a requisite role in regulating the lethal effects of anaphylatoxin-mediated shock, 2) that these lethal effects are mediated predominantly by C5a-induced histamine release, and 3) that C3a does not contribute significantly to shock following acute complement activation.
Collapse
Affiliation(s)
- Stacey L Mueller-Ortiz
- Research Center for Immunology and Autoimmune Diseases, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
34
|
Chimote G, Banerjee R. Evaluation of antitubercular drug-loaded surfactants as inhalable drug-delivery systems for pulmonary tuberculosis. J Biomed Mater Res A 2009; 89:281-92. [PMID: 18431766 DOI: 10.1002/jbm.a.31959] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pulmonary tuberculosis is associated with a year-long chemotherapy, poor alveolar drug levels, drug-related systemic toxicity, and patient noncompliance. In this study, exogenous pulmonary surfactant is proposed as a drug carrier for antitubercular drugs. Dipalmitoylphosphatidylcholine (DPPC), the major lung-surfactant lipid, has been combined with antitubercular drugs isoniazid (INH), rifampicin (RFM), and ethambutol (ETH) in 1:1 ratio by weight, in which drugs had a ratio of 1:2:3 by weight. At 37 degrees C, the formulation had better surfactant function with quicker reduction of surface tension on adsorption (32.71 +/- 0.65 mN/m) than DPPC liposomes (44.67 +/- 0.57 mN/m) and maintained 100% airway patency in a capillary surfactometer. Drug-loaded surfactant liposomes were about 2 microm and had entrapment efficiency of 30.04% +/- 2.05%, 18.85% +/- 2.92%, and 61.47% +/- 3.32% for INH, RFM, and ETH, respectively. Sustained release of the drugs from surfactants was observed over 24 h. In vitro alveolar deposition efficiency using the twin impinger showed 12.06% +/- 1.87% of INH, 43.30% +/- 0.87% of RFM, and 22.07% +/- 2.02% of ETH deposited in the alveolar chamber upon nebulization for a minute using a jet nebulizer. The formulation was biocompatible and stable with physicochemical properties being retained even after storage for a month at 4 degrees C. Antitubercular drug-loaded surfactants developed could serve dual purposes of alveolar stabilization due to surfactant action and better reach of these drugs to the alveoli due to antiatelectatic effect of the surfactant.
Collapse
Affiliation(s)
- G Chimote
- School of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400 076, India
| | | |
Collapse
|
35
|
Kaplan AP, Joseph K, Maykut RJ, Geba GP, Zeldin RK. Treatment of chronic autoimmune urticaria with omalizumab. J Allergy Clin Immunol 2008; 122:569-73. [PMID: 18774392 DOI: 10.1016/j.jaci.2008.07.006] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Revised: 06/20/2008] [Accepted: 07/03/2008] [Indexed: 10/21/2022]
Abstract
BACKGROUND Approximately 45% of patients with chronic urticaria have an IgG autoantibody directed to the alpha-subunit of the high-affinity IgE receptor (chronic autoimmune urticaria, CAU) leading to cutaneous mast cell and basophil activation. Treatment of allergic asthma with omalizumab produces rapid reduction in free IgE levels and subsequent decrease in Fc epsilon RI expression on mast cells and basophils. If this occurs in CAU, cross-linking of IgE receptors by autoantibody would be less likely, reducing cell activation and urticaria/angioedema. OBJECTIVE To investigate the efficacy of omalizumab in patients with CAU symptomatic despite antihistamine therapy. METHODS Twelve patients with CAU, identified by basophil histamine release assay and autologous skin test, with persistent symptoms for at least 6 weeks despite antihistamines, were treated with placebo for 4 weeks followed by omalizumab (>or=0.016 mg/kg/IU mL(-1) IgE per month) every 2 or 4 weeks for 16 weeks. Primary efficacy variable was change from baseline to the final 4 weeks of omalizumab treatment in mean Urticaria Activity Score (UAS, 0-9 scale). Changes in rescue medication use and quality of life were assessed. RESULTS Mean UAS declined significantly from baseline to the final 4 weeks of omalizumab treatment (7.50 +/- 1.78 to 2.66 +/- 3.31, -4.84 +/- 2.86, P = .0002). Seven patients achieved complete symptom resolution. In 4 patients, mean UAS decreased, but urticaria persisted. One patient did not respond. Rescue medication use was reduced significantly, and quality of life improved. No adverse effects were reported or observed. CONCLUSION This exploratory proof of concept study suggests omalizumab is an effective therapy for CAU resistant to antihistamines.
Collapse
Affiliation(s)
- Allen P Kaplan
- National Allergy, Asthma, and Urticaria Centers of Charleston, Charleston, SC, USA.
| | | | | | | | | |
Collapse
|
36
|
Chang JY, Lin CCJ, Salamanca S, Pangburn MK, Wetsel RA. Denaturation and unfolding of human anaphylatoxin C3a: an unusually low covalent stability of its native disulfide bonds. Arch Biochem Biophys 2008; 480:104-10. [PMID: 18854167 DOI: 10.1016/j.abb.2008.09.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Accepted: 09/22/2008] [Indexed: 11/19/2022]
Abstract
The complement C3a anaphylatoxin is a major molecular mediator of innate immunity. It is a potent activator of mast cells, basophils and eosinophils and causes smooth muscle contraction. Structurally, C3a is a relatively small protein (77 amino acids) comprising a N-terminal domain connected by 3 native disulfide bonds and a helical C-terminal segment. The structural stability of C3a has been investigated here using three different methods: Disulfide scrambling; Differential CD spectroscopy; and Reductive unfolding. Two uncommon features regarding the stability of C3a and the structure of denatured C3a have been observed in this study. (a) There is an unusual disconnection between the conformational stability of C3a and the covalent stability of its three native disulfide bonds that is not seen with other disulfide proteins. As measured by both methods of disulfide scrambling and differential CD spectroscopy, the native C3a exhibits a global conformational stability that is comparable to numerous proteins with similar size and disulfide content, all with mid-point denaturation of [GdmCl](1/2) at 3.4-5M. These proteins include hirudin, tick anticoagulant protein and leech carboxypeptidase inhibitor. However, the native disulfide bonds of C3a is 150-1000 fold less stable than those proteins as evaluated by the method of reductive unfolding. The 3 native disulfide bonds of C3a can be collectively and quantitatively reduced with as low as 1mM of dithiothreitol within 5 min. The fragility of the native disulfide bonds of C3a has not yet been observed with other native disulfide proteins. (b) Using the method of disulfide scrambling, denatured C3a was shown to consist of diverse isomers adopting varied extent of unfolding. Among them, the most extensively unfolded isomer of denatured C3a is found to assume beads-form disulfide pattern, comprising Cys(36)-Cys(49) and two disulfide bonds formed by two pair of consecutive cysteines, Cys(22)-Cys(23) and Cys(56)-Cys(57), a unique disulfide structure of polypeptide that has not been documented previously.
Collapse
Affiliation(s)
- Jui-Yoa Chang
- Research Center for Protein Chemistry, Institute of Molecular Medicine, The University of Texas Medical School, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
37
|
Nemali S, Siemsen DW, Nelson LK, Bunger PL, Faulkner CL, Rainard P, Gauss KA, Jutila MA, Quinn MT. Molecular analysis of the bovine anaphylatoxin C5a receptor. J Leukoc Biol 2008; 84:537-49. [PMID: 18480166 DOI: 10.1189/jlb.0208142] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recruitment of phagocytes to inflammatory sites involves the coordinated action of several chemoattractants, including the anaphylatoxin C5a. While the C5a receptor (C5aR) has been well characterized in humans and rodents, little is known about the bovine C5aR. Here, we report cloning of bovine C5R1, the gene encoding bovine C5aR. We also analyzed genomic sequence upstream of the C5R1 translation start site. Although the bovine C5aR amino acid sequence was well conserved among species, significant differences in conserved features were found, including major differences in the N terminus, intracellular loop 3, and transmembrane domain VII. Analysis of C5aR expression by flow cytometry and confocal microscopy demonstrated high levels of C5aR on all bovine neutrophils and a subset of bovine monocytes. C5aR was not expressed on resting or activated bovine lymphocytes, although C5aR message was present in these cells. C5aR was also expressed on a small subset of bovine mammary epithelial cells. Pharmacological analysis of bovine C5aR-mediated responses showed that bovine C5a and C5adesArg both induced dose-dependent calcium fluxes and chemotaxis in bovine neutrophils, with similar efficacy for both agonists. Treatment of bovine neutrophils with C5a or C5adesArg resulted in homologous desensitization of bovine C5aR and cross-desensitization to interleukin 8 (IL-8) and platelet-activating factor (PAF); whereas, treatment with IL-8 or PAF did not cross-desensitize the cells to C5a or C5adesArg. Overall, these studies provide important information regarding distinct structural and functional features that may contribute to the unique pharmacological properties of bovine C5aR.
Collapse
Affiliation(s)
- Sailasree Nemali
- Department of Veterinary Molecular Biology, Montana State University, Bozeman, MT 59717, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
A conformationally-biased, response-selective agonist of C5a acts as a molecular adjuvant by modulating antigen processing and presentation activities of human dendritic cells. Int Immunopharmacol 2008; 8:819-27. [PMID: 18442785 DOI: 10.1016/j.intimp.2008.01.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Revised: 01/28/2008] [Accepted: 01/30/2008] [Indexed: 11/22/2022]
Abstract
A partial mechanism by which a conformationally-biased, response-selective agonist of complement component C5a, Tyr-Ser-Phe-Lys-Pro-Met-Pro-Leu-D-Ala-Arg or YSFKPMPLaR (EP54), acts as a molecular adjuvant is presented by showing the manner in which this peptide engages human dendritic cells (DC). Confocal microscopy was used to show that fluorescent-labeled EP54 (0.2 microM) and fluorescent-labeled B and T cell epitopes attached to EP54 (i.e., EP54-containing vaccines, 0.2 microM) were internalized by human DCs well within 30 min of exposure. After 24 h of exposure, EP54 and the B and T cell epitopes of the EP54-containing vaccines (20 microM) were presented on the DC surface in the context of HLA-ABC and HLA-DR determinants. Also, exposure of DCs to EP54 (50 microg/ml) induced the activation of genes specific for the Th1 cytokines IL-6, IL-12, INFgamma, and TNFalpha as well as the Th2 cytokine IL-4. Internalization, HLA expression, and cytokine gene activation were not observed in the presence of the inactive, scrambled EP54 constructs arguing that these effects of EP54 are mediated predominately via C5a receptors on the DC surface.
Collapse
|
39
|
Tse PK, Lee YL, Chow WN, Luk JMC, Lee KF, Yeung WSB. Preimplantation embryos cooperate with oviductal cells to produce embryotrophic inactivated complement-3b. Endocrinology 2008; 149:1268-76. [PMID: 18039777 DOI: 10.1210/en.2007-1277] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human oviductal epithelial (OE) cells produce complement protein 3 (C3) and its derivatives, C3b and inactivated complement-3b (iC3b). Among them, iC3b is the most potent embryotrophic molecule. We studied the production of iC3b in the oviductal cell/embryo culture system. In the immune system, C3 convertase converts C3 into C3b, and the conversion of C3b to iC3b requires factor I (fI) and its cofactors, such as factor H or membrane cofactor protein. Human oviductal epithelium and OE cells expressed mRNA and protein of the components of C3 convertase, including C2, C4, factor B, and factor D. The OE cell-conditioned medium contained active C3 convertase activity that was suppressed by C3 convertase inhibitor, H17 in a dose and time-dependent manner. Although the oviductal epithelium and OE cells produced fI, the production of its cofactor, factor H required for the conversion of C3b to iC3b, was weak. Thus, OE cell-conditioned medium was inefficient in producing iC3b from exogenous C3b. On the contrary, mouse embryos facilitated such conversion to iC3b, which was taken up by the embryos, resulting in the formation of more blastocysts of larger size. The facilitatory activity was mediated by complement receptor 1-related gene/protein Y (Crry) with known membrane cofactor protein activity on the trophectoderm of the embryos as anti-Crry antibody inhibited the conversion and embryotrophic activity of C3b in the presence of fI. In conclusion, human oviduct possesses C3 convertase activity converting C3 to C3b, and Crry of the preimplantation embryos may be involved in the production of embryotrophic iC3b on the surface of the embryos.
Collapse
Affiliation(s)
- Pui-Keung Tse
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, China
| | | | | | | | | | | |
Collapse
|
40
|
Lee H, Whitfeld PL, Mackay CR. Receptors for complement C5a. The importance of C5aR and the enigmatic role of C5L2. Immunol Cell Biol 2008; 86:153-60. [PMID: 18227853 DOI: 10.1038/sj.icb.7100166] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Complement component C5a is one of the most potent inflammatory chemoattractants and has been implicated in the pathogenesis of numerous inflammatory diseases. C5a binds two receptors, C5aR and C5L2. Most of the C5a functional effects occur through C5aR, and the pharmaceutical industry has focused on this receptor for the development of new anti-inflammatory therapies. We used a novel approach to generate and test therapeutics that target C5aR. We created human C5aR knock-in mice, and used neutrophils from these to immunize wild-type mice. This yielded high-affinity blocking mAbs to human C5aR. We tested these anti-human C5aR mAbs in mouse models of inflammation, using the human C5aR knock-in mice. These antibodies completely prevented disease onset and were also able to reverse established disease in the K/B x N arthritis model. The physiological role of the other C5a receptor, C5L2 is still unclear, and our studies with blocking mAbs to human C5L2 have failed to demonstrate a clear functional role in signaling to C5a. The development of effective mAbs to human C5aR is an alternative approach to drug development, for this highly attractive target.
Collapse
Affiliation(s)
- Hyun Lee
- Immunology and Inflammation Department, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | | | | |
Collapse
|
41
|
Haupt MT. Anaphylaxis and Anaphylactic Shock. Crit Care Med 2008. [DOI: 10.1016/b978-032304841-5.50031-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
42
|
Hollmann TJ, Mueller-Ortiz SL, Braun MC, Wetsel RA. Disruption of the C5a receptor gene increases resistance to acute Gram-negative bacteremia and endotoxic shock: opposing roles of C3a and C5a. Mol Immunol 2007; 45:1907-15. [PMID: 18063050 DOI: 10.1016/j.molimm.2007.10.037] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Revised: 10/24/2007] [Accepted: 10/24/2007] [Indexed: 11/15/2022]
Abstract
The host response to intravascular, Gram-negative bacteria includes profound immunologic, hematologic and physiologic changes. Numerous host defense mechanisms are activated by Gram-negative bacteria, including the complement system. Activation of the complement system leads to cleavage of C5 with subsequent generation of the C5a anaphylatoxin peptide. C5a mediates potent, proinflammatory activities by binding to the C5a receptor (C5aR, CD88). In this study, we report the targeted disruption of the murine C5aR gene (C5aR-/- mice) and define the role of the C5aR in a model of Gram-negative bacteremia. Following an intravenous infusion of heat-killed Escherichia coli, the C5aR-/- mice were completely protected from the mortality suffered by their wild-type littermates (P<0.001). The C5aR-/- mice were also significantly (P=0.008) more resistant to mortality following an intravenous infusion of purified E. coli endotoxin compared to the wild-type littermates. In addition, the C5aR-/- mice were resistant to the thrombocytopenia and hemoconcentration observed in wild-type animals. Lethality in the wild-type mice was reversed by pre-treatment with either the histamine antagonist diphenhydramine or triprolidine. The wild-type littermates were also rescued following pre-treatment with the basophil and mast cell-stabilizing agent - cromolyn sodium. Collectively, these data demonstrate that not only is the absence of the C5aR protective in E. coli bacteremia, but that C5aR-dependent histamine release plays a major role in shock induced by Gram-negative septicemia. Moreover, they provide additional in vivo evidence that C3a and C5a have divergent biological functions in Gram-negative bacteremia and shock.
Collapse
Affiliation(s)
- Travis J Hollmann
- Research Center for Immunology and Autoimmune Diseases, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas-Houston, TX77030, USA
| | | | | | | |
Collapse
|
43
|
Skidgel RA, Erdös EG. Structure and function of human plasma carboxypeptidase N, the anaphylatoxin inactivator. Int Immunopharmacol 2007; 7:1888-99. [PMID: 18039526 DOI: 10.1016/j.intimp.2007.07.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2007] [Accepted: 07/06/2007] [Indexed: 12/27/2022]
Abstract
Human carboxypeptidase N (CPN) was discovered in the early 1960s as a plasma enzyme that inactivates bradykinin and was identified 8 years later as the major "anaphylatoxin inactivator" of blood. CPN plays an important role in protecting the body from excessive buildup of potentially deleterious peptides that normally act as local autocrine or paracrine hormones. This review summarizes the structure, enzymatic properties and function of this important human enzyme, including insights gained by the recent elucidation of the crystal structure of the CPN catalytic subunit and structural modeling of the non-catalytic regulatory 83 kDa subunit. We also discuss its physiological role in cleaving substrates such as kinins, anaphylatoxins, creatine kinase, plasminogen receptors, hemoglobin and stromal cell-derived factor-1alpha (SDF-1alpha).
Collapse
Affiliation(s)
- Randal A Skidgel
- Department of Pharmacology, University of Illinois at Chicago College of Medicine Chicago, IL 60612, USA.
| | | |
Collapse
|
44
|
Higuchi S, Murayama N, Saguchi KI, Ohi H, Fujita Y, da Silva NJ, de Siqueira RJB, Lahlou S, Aird SD. A novel peptide from the ACEI/BPP-CNP precursor in the venom of Crotalus durissus collilineatus. Comp Biochem Physiol C Toxicol Pharmacol 2006; 144:107-21. [PMID: 16979945 DOI: 10.1016/j.cbpc.2006.04.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2006] [Revised: 04/17/2006] [Accepted: 04/19/2006] [Indexed: 10/24/2022]
Abstract
In crotaline venoms, angiotensin-converting enzyme inhibitors [ACEIs, also known as bradykinin potentiating peptides (BPPs)], are products of a gene coding for an ACEI/BPP-C-type natriuretic peptide (CNP) precursor. In the genes from Bothrops jararaca and Gloydius blomhoffii, ACEI/BPP sequences are repeated. Sequencing of a cDNA clone from venom glands of Crotalus durissus collilineatus showed that two ACEIs/BPPs are located together at the N-terminus, but without repeats. An additional sequence for CNP was unexpectedly found at the C-terminus. Homologous genes for the ACEI/BPP-CNP precursor suggest that most crotaline venoms contain both ACEIs/BPPs and CNP. The sequence of ACEIs/BPPs is separated from the CNP sequence by a long spacer sequence. Previously, there was no evidence that this spacer actually coded any expressed peptides. Aird and Kaiser (1986, unpublished) previously isolated and sequenced a peptide of 11 residues (TPPAGPDVGPR) from Crotalus viridis viridis venom. In the present study, analysis of the cDNA clone from C. d. collilineatus revealed a nearly identical sequence in the ACEI/BPP-CNP spacer. Fractionation of the crude venom by reverse phase HPLC (C(18)), and analysis of the fractions by mass spectrometry (MS) indicated a component of 1020.5 Da. Amino acid sequencing by MS/MS confirmed that C. d. collilineatus venom contains the peptide TPPAGPDGGPR. Its high proline content and paired proline residues are typical of venom hypotensive peptides, although it lacks the usual N-terminal pyroglutamate. It has no demonstrable hypotensive activity when injected intravenously in rats; however, its occurrence in the venoms of dissimilar species suggests that its presence is not accidental. Evidence suggests that these novel toxins probably activate anaphylatoxin C3a receptors.
Collapse
Affiliation(s)
- Shigesada Higuchi
- Showa University School of Pharmaceutical Sciences, Shinagawa-ku, Tokyo 142-8555, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
TERAN LM, CAMPOS MG, BEGISHVILLI BT, SCHRÖDER JM, DJUKANOVIC R, SHUTE JK, CHURCH MK, HOLGATE ST, DAVIES DE. Identification of neutrophil chemotactie factors in bronchoalveolar lavage fluid of asthmatic patients. Clin Exp Allergy 2006. [DOI: 10.1111/j.1365-2222.1997.tb00724.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
46
|
Bhakdi S, Tranum-Jensen J. Damage to mammalian cells by proteins that form transmembrane pores. Rev Physiol Biochem Pharmacol 2005; 107:147-223. [PMID: 3303271 DOI: 10.1007/bfb0027646] [Citation(s) in RCA: 202] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
47
|
Waters SM, Brodbeck RM, Steflik J, Yu J, Baltazar C, Peck AE, Severance D, Zhang LY, Currie K, Chenard BL, Hutchison AJ, Maynard G, Krause JE. Molecular characterization of the gerbil C5a receptor and identification of a transmembrane domain V amino acid that is crucial for small molecule antagonist interaction. J Biol Chem 2005; 280:40617-23. [PMID: 16230349 DOI: 10.1074/jbc.m509245200] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Anaphylatoxin C5a is a potent inflammatory mediator associated with pathogenesis and progression of several inflammation-associated disorders. Small molecule C5a receptor (C5aR) antagonist development is hampered by species-specific receptor biology and the associated inability to use standard rat and mouse in vivo models. Gerbil is one rodent species reportedly responsive to small molecule C5aR antagonists with human C5aR affinity. We report the identification of the gerbil C5aR cDNA using a degenerate primer PCR cloning strategy. The nucleotide sequence revealed an open reading frame encoding a 347-amino acid protein. The cloned receptor (expressed in Sf9 cells) bound recombinant human C5a with nanomolar affinity. Alignment of the gerbil C5aR sequence with those from other species showed that a Trp residue in transmembrane domain V is the only transmembrane domain amino acid unique to small molecule C5aR antagonist-responsive species (i.e. gerbil, human, and non-human primate). Site-directed mutagenesis was used to generate human and mouse C5aRs with a residue exchange of this Trp residue. Mutation of Trp to Leu in human C5aR completely eliminated small molecule antagonist-receptor interaction. In contrast, mutation of Leu to Trp in mouse C5aR enabled small molecule antagonist-receptor interaction. This crucial Trp residue is located deeper within transmembrane domain V than residues reportedly involved in C5a- and cyclic peptide C5a antagonist-receptor interaction, suggesting a novel interaction site(s) for small molecule antagonists. These data provide insight into the basis for small molecule antagonist species selectivity and further define sites critical for C5aR activation and function.
Collapse
Affiliation(s)
- Stephen M Waters
- Department of Biochemistry and Molecular Biology, Neurogen Corporation, Branford, Connecticut 06405, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Valenzuela JG. Exploring tick saliva: from biochemistry to ‘sialomes’ and functional genomics. Parasitology 2005; 129 Suppl:S83-94. [PMID: 15938506 DOI: 10.1017/s0031182004005189] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Tick saliva, a fluid once believed to be only relevant for lubrication of mouthparts and water balance, is now well known to be a cocktail of potent anti-haemostatic, anti-inflammatory and immunomodulatory molecules that helps these arthropods obtain a blood meal from their vertebrate hosts. The repertoire of pharmacologically active components in this cocktail is impressive as well as the number of targets they specifically affect. These salivary components change the physiology of the host at the bite site and, consequently, some pathogens transmitted by ticks take advantage of this change and become more infective. Tick salivary proteins have therefore become an attractive target to control tick-borne diseases. Recent advances in molecular biology, protein chemistry and computational biology are accelerating the isolation, sequencing and analysis of a large number of transcripts and proteins from the saliva of different ticks. Many of these newly isolated genes code for proteins with homologies to known proteins allowing identification or prediction of their function. However, most of these genes code for proteins with unknown functions therefore opening the road to functional genomic approaches to identify their biological activities and roles in blood feeding and hence, vaccine development to control tick-borne diseases.
Collapse
Affiliation(s)
- J G Valenzuela
- Vector Molecular Biology Unit, Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, 4 Center Drive, 4/B2-35, Bethesda, MD 20892, USA.
| |
Collapse
|
49
|
Glovsky MM, Ward PA, Johnson KJ. Complement determinations in human disease. Ann Allergy Asthma Immunol 2005; 93:513-22; quiz 523-5, 605. [PMID: 15609759 DOI: 10.1016/s1081-1206(10)61257-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To define techniques used for complement measurements and examine the clinical relevance of alterations of complement determinations in disease. DATA SOURCES Data have been assembled from the authors' research, original articles, and reviews, as well as chapters and complete books on complement. STUDY SELECTION Studies were chosen for inclusion by the opinions of the authors, relevant complement reviews, publications, and books. RESULTS Complement has been shown to possess approximately 31 proteins, some of which are enzymes (C1r, C1s, C2, factor B, factor D), some cofactors, some inhibitors or inactivators, and others composed of membrane-integrated proteins. All of the complement proteins have been purified, and many of the respective genes have been identified. The complement cascade is a dual-edged sword, causing protection against bacterial and viral invasion by promoting phagocytosis and inflammation. Pathologically, complement can cause substantial damage to blood vessels (vasculitis), kidney basement membrane and attached endothelial and epithelial cells (nephritis), joint synovium (arthritis), and erythrocytes (hemolysis) if it is not adequately controlled. CONCLUSIONS Definitive evidence is available that complement-mediated tissue destruction occurs after immune complex injury in the kidney and lung and may be important in lupus erythematosus and adult respiratory distress syndrome. Future studies on complement receptor structure and function may provide clues to treat effectively lupus, hemolytic anemias, and nephritis. In addition, gene therapy and antibody therapy need further refinement to treat immunodeficiency diseases.
Collapse
|
50
|
Braun MC, Reins RY, Li TB, Hollmann TJ, Dutta R, Rick WA, Teng BB, Ke B. Renal expression of the C3a receptor and functional responses of primary human proximal tubular epithelial cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:4190-6. [PMID: 15356170 DOI: 10.4049/jimmunol.173.6.4190] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although complement activation and deposition have been associated with a variety of glomerulopathies, the pathogenic mechanisms by which complement directly mediates renal injury remain to be fully elucidated. Renal parenchymal tissues express a limited repertoire of receptors that directly bind activated complement proteins. We report the renal expression of the receptor for the C3 cleavage product C3a, a member of the anaphylatoxin family. C3aR is highly expressed in normal human and murine kidney, as demonstrated by immunohistochemistry and in situ hybridization. Its distribution is limited to epithelial cells only, as glomerular endothelial and mesangial cells showed no evidence of C3aR expression. The C3aR is also expressed by primary renal proximal tubular epithelial cells in vitro as demonstrated by FACS, Western blot, and RT-PCR. In vitro C3aR is functional in terms of its capacity to bind 125I-labeled C3a and generate inositol triphosphate. Finally, using microarray analysis, four novel genes were identified and confirmed as transcriptionally regulated by C3aR activation in proximal tubular cells. These studies define a new pathway by which complement activation may directly modulate the renal response to immunologic injury.
Collapse
MESH Headings
- Animals
- Complement Activation/genetics
- Complement Activation/immunology
- Complement C3a/metabolism
- Epithelial Cells/immunology
- Epithelial Cells/metabolism
- Epithelial Cells/physiology
- Expressed Sequence Tags
- Gene Expression Profiling
- Humans
- Immunohistochemistry
- In Situ Hybridization
- Kidney Glomerulus/cytology
- Kidney Glomerulus/immunology
- Kidney Glomerulus/metabolism
- Kidney Glomerulus/physiology
- Kidney Tubules, Proximal/cytology
- Kidney Tubules, Proximal/immunology
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/physiology
- Membrane Proteins/biosynthesis
- Membrane Proteins/deficiency
- Membrane Proteins/genetics
- Mice
- Mice, Knockout
- Oligonucleotide Array Sequence Analysis
- RNA, Messenger/biosynthesis
- Receptors, Complement/biosynthesis
- Receptors, Complement/deficiency
- Receptors, Complement/genetics
- Transcription, Genetic/immunology
Collapse
Affiliation(s)
- Michael C Braun
- Center for Immunology, Brown Foundation Institute of Molecular Medicine, University of Texas, Houston 77030, USA.
| | | | | | | | | | | | | | | |
Collapse
|