Go to JCI Insight
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
  • Clinical Research and Public Health
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Gastroenterology
    • Immunology
    • Metabolism
    • Nephrology
    • Neuroscience
    • Oncology
    • Pulmonology
    • Vascular biology
    • All ...
  • Videos
    • Conversations with Giants in Medicine
    • Video Abstracts
  • Reviews
    • View all reviews ...
    • Pancreatic Cancer (Jul 2025)
    • Complement Biology and Therapeutics (May 2025)
    • Evolving insights into MASLD and MASH pathogenesis and treatment (Apr 2025)
    • Microbiome in Health and Disease (Feb 2025)
    • Substance Use Disorders (Oct 2024)
    • Clonal Hematopoiesis (Oct 2024)
    • Sex Differences in Medicine (Sep 2024)
    • View all review series ...
  • Viewpoint
  • Collections
    • In-Press Preview
    • Clinical Research and Public Health
    • Research Letters
    • Letters to the Editor
    • Editorials
    • Commentaries
    • Editor's notes
    • Reviews
    • Viewpoints
    • 100th anniversary
    • Top read articles

  • Current issue
  • Past issues
  • Specialties
  • Reviews
  • Review series
  • Conversations with Giants in Medicine
  • Video Abstracts
  • In-Press Preview
  • Clinical Research and Public Health
  • Research Letters
  • Letters to the Editor
  • Editorials
  • Commentaries
  • Editor's notes
  • Reviews
  • Viewpoints
  • 100th anniversary
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
Top
  • View PDF
  • Download citation information
  • Send a comment
  • Terms of use
  • Standard abbreviations
  • Need help? Email the journal
  • Top
  • Abstract
  • Introduction
  • Animal testing in biomedical research
  • Using animals to understand the human complement system
  • Evaluating complement-targeted therapies: animal models versus clinical trials
  • Discussion and remarks
  • Supplemental material
  • Acknowledgments
  • Footnotes
  • References
  • Version history
  • Article usage
  • Citations to this article

Advertisement

Review Series Open Access | 10.1172/JCI188347

Friend or foe: assessing the value of animal models for facilitating clinical breakthroughs in complement research

Felix Poppelaars, V. Michael Holers, and Joshua M. Thurman

Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA.

Address correspondence to: Felix Poppelaars, Division of Nephrology and Hypertension, University of Colorado School of Medicine, B-115 1775 Aurora Court, M20-3103, Aurora, Colorado 80045, USA. Email: felix.poppelaars@cuanschutz.edu. Or to: Joshua M. Thurman, Division of Nephrology and Hypertension, University of Colorado School of Medicine, B-115 1775 Aurora Court, M20-3103, Aurora, Colorado 80045, USA. Email: joshua.thurman@cuanschutz.edu.

Find articles by Poppelaars, F. in: PubMed | Google Scholar

Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA.

Address correspondence to: Felix Poppelaars, Division of Nephrology and Hypertension, University of Colorado School of Medicine, B-115 1775 Aurora Court, M20-3103, Aurora, Colorado 80045, USA. Email: felix.poppelaars@cuanschutz.edu. Or to: Joshua M. Thurman, Division of Nephrology and Hypertension, University of Colorado School of Medicine, B-115 1775 Aurora Court, M20-3103, Aurora, Colorado 80045, USA. Email: joshua.thurman@cuanschutz.edu.

Find articles by Holers, V. in: PubMed | Google Scholar

Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA.

Address correspondence to: Felix Poppelaars, Division of Nephrology and Hypertension, University of Colorado School of Medicine, B-115 1775 Aurora Court, M20-3103, Aurora, Colorado 80045, USA. Email: felix.poppelaars@cuanschutz.edu. Or to: Joshua M. Thurman, Division of Nephrology and Hypertension, University of Colorado School of Medicine, B-115 1775 Aurora Court, M20-3103, Aurora, Colorado 80045, USA. Email: joshua.thurman@cuanschutz.edu.

Find articles by Thurman, J. in: PubMed | Google Scholar |

Published June 16, 2025 - More info

Published in Volume 135, Issue 12 on June 16, 2025
J Clin Invest. 2025;135(12):e188347. https://doi.org/10.1172/JCI188347.
© 2025 Poppelaars et al. This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
Published June 16, 2025 - Version history
View PDF
Abstract

Animal experiments have long been a cornerstone of advancements in biomedical research, particularly in developing novel therapeutic strategies for inflammatory and autoimmune diseases. However, these historically important approaches are now facing growing scrutiny for ethical reasons, concerns about translational limitations to human biology, and the rising availability of animal-free research methods. This shift raises a critical question: How relevant and effective are animal models for driving future advancements in today’s research landscape? This Review aims to explore this question within the field of biomedical research on the complement system, critically evaluating the contribution of animal models to the recent advancements and clinical successes of complement-targeted therapies. Specifically, we assess areas where animal studies have been indispensable for elucidating disease mechanisms and conducting preclinical evaluations, alongside instances where findings from animal models failed to translate successfully to human trials. Furthermore, we discuss similarities and differences in the complement system between animals and humans and explore innovations in animal research designed to improve translatability to human biology. By assessing the contributions of animal studies to complement therapeutics, this Review aims to provide insights into animal models’ strengths, limitations, and evolving role in complement research.

Introduction

The complement system is a network of circulating and membrane-bound proteins (Tables 1, 2, 3, 4, and 5), crucial for immune defense and tissue homeostasis, while driving inflammation and injury during disease (1). As an ancient component of immunity, complement is highly conserved across vertebrates, with primitive forms present even in invertebrates (2). Research on this system, utilizing in vitro assays, animal models, and human samples, has provided key insights into its biology and role in numerous diseases (3). The FDA has currently approved 11 complement inhibitors for 11 disease indications (Supplemental Table 1; supplemental material available online with this article; https://doi.org/10.1172/JCI188347DS1), with more under investigation (4). Although clinical complement inhibition has only recently gained momentum, animal models have demonstrated its therapeutic potential for over 50 years (1, 5). However, the use of animal models is increasingly questioned because of ethical and translational concerns. For anti–neutrophil cytoplasmic antibody–associated vasculitis (AAV), animal models were crucial in uncovering the role of complement in its pathogenesis and achieving approval of the C5a receptor antagonist in this disease (6, 7). In contrast, complement inhibitors for paroxysmal nocturnal hemoglobinuria (PNH) were approved almost entirely based on studies using human blood samples (8, 9). Presently, the clinical use of complement therapeutics enables the direct study of complement inhibition in humans (10). This, combined with innovations in molecular techniques and tools — i.e., large-scale genomics (11, 12), AI-assisted methodology (13, 14), and organoids (15) — raises an important question in complement research: How essential is it to continue conducting animal experiments to achieve future success?

Table 1

The initiators of the classical pathway and lectin pathway in mice and humans

Table 2

Other proteins related to the classical pathway and lectin pathway in mice and humans

Table 3

The AP in mice and humans

Table 4

Complement regulators and complement receptors in mice and humans

Table 5

The terminal pathway and anaphylatoxin receptors in mice and humans

Although quantifying the contribution of animal models to clinical advancements in the complement field is challenging, this Review aims to assess areas where animal studies have made substantial contributions and where they have fallen short or proved unnecessary.

Animal testing in biomedical research

The rise of animal-free methods, along with concerns about animal rights, ethics, high costs, and translatability, sparked skepticism regarding the continued reliance on animal testing. This shift is reflected in policy changes in the United States (Frank R. Lautenberg Chemical Safety Act — Toxic Substances Control Act) and the European Union (Registration, Evaluation, Authorisation and Restriction of Chemicals Regulation & 223/2009 EU CPR), placing animal research under heightened scrutiny. Additionally, the FDA now permits nonanimal testing alternatives for instances like biosimilar drugs and toxicity (FDA Modernization Act 2.0 — S.5002). Surveys indicate declining public support for animal testing and growing preference for its elimination (16). However, negative media coverage and misinformation contribute to unfavorable perceptions of animal testing (17), while increased awareness of regulations protecting laboratory animals improves attitudes (18). This complicates the determination of whether current negative views stem from misinformation or informed opinions.

Animal testing in research is under strict regulations in North America, the European Union, and several other countries, based on the principles of the 3 Rs (Replacement, Reduction, and Refinement) (summarized in ref. 19). Established over 50 years ago, these principles provide a framework for conducting (more) ethical animal research. In brief, Reduction involves using the minimum number of animals needed for reliable results, while Refinement focuses on minimizing pain and suffering. Replacement entails employing nonanimal methods, when possible, either absolutely or relatively (animals provide organs or tissues for in vitro experiments). Replacement can also involve substituting vertebrates with species that have a reduced capacity to feel pain (i.e., invertebrates or bacteria). Other strategies to reduce animal use include improved study design, method development, and project coordination. In silico (computational modeling), in vitro, and ex vivo approaches can also support the Reduction and Replacement principles.

Using animals to understand the human complement system

Despite being highly conserved across vertebrates, notable differences still exist between humans and research animals (Tables 1–5) (20). Animal models are selected for their ability to standardize and manipulate, thereby determining causality. Furthermore, research in intact organisms provides context, as the complement system operates in circulation and locally in tissues, while interacting with other systems (1, 3). Animal studies provided valuable insights, but not all findings translate to humans (3, 21). What, then, makes a model suitable for complement research, particularly for developing diagnostics and therapies for human diseases? Besides anatomical, physiological, and disease-related similarities, it is crucial to evaluate aspects of the complement system relevant to the research question, including phylogenetic proximity, sequence alignment, structure, functionality, protein interactions, and expression levels.

Commonalities in the complement system across species. Mammals, birds, amphibians, and fish generally possess a complete set of complement genes, with few exceptions (2). C3, a central component of the complement cascade, shows strong conservation across species (Figure 1) (22, 23). Similarly, there is a high degree of interspecies amino acid sequence homology with Factor B, along with collectin-10 and collectin-11 of the lectin pathway (LP) (2, 20, 24). Even though Factor H, a soluble complement regulator, is only 63% identical between humans and mice, the structural organization and functional roles remain highly similar (25). In primitive invertebrates, C3-like molecules retain key structural features analogous to human C3, including the thioester moiety (which enables covalent binding to surfaces), anaphylatoxin domain (C3a fragment), cleavage site (forming C3b), and Factor B binding site (C3-convertase assembly) (22, 23). Furthermore, many complement proteins exhibit functional cross-species reactivity (26). Importantly, protein-protein interactions in the complement system are highly conserved across species, such as C1q with IgG (27), MASPs with mannose-binding lectin (28), Factor H with C3d (25), and CD59 with C8 (29). Reduced homology and loss of cross-reactivity with human counterparts can result from coevolution to preserve key protein interactions, maintaining the fundamental framework of the complement system within species (29). Together, these functional similarities underscore the value of animal models in studying the complement system’s role in human diseases.

Conservation of structure and function of C3 across species.Figure 1

Conservation of structure and function of C3 across species. (A) C3 is the central and most abundant circulating complement protein, forming the pivotal convergence point of all pathways. (B) Phylogenetic tree illustrating the early emergence of C3-like genes in primitive invertebrates. (C) Human C3 protein (UniProt Knowledgebase [UniProtKB]: P01024) exhibits significant homology with other animals, including mouse (UniProtKB: P01027), rat (UniProtKB: P01026), guinea pig (UniProtKB: P12387), pig (UniProtKB: P01025), and cynomolgus monkey (UniProtKB: A0A2K6D5R0). Percentages represent amino acid identity shared with human C3 and were obtained using the Align function on UniProt. (D) Human C3 consists of eight macroglobulin domains (MG1–MG8); an ANA domain; a linker (LNK) domain; a C1r/s, Uegf, B (CUB) domain; a TED; and a C345C domain. Structure of human C3 adapted from Zarantonello et al. with permission (368). Moreover, functional characteristics of C3 are conserved even in the most primitive invertebrates: (i) Cleavage of C3 removes the ANA domain (forming C3a) and induces conformational changes (forming C3b), exposing the reactive TED that enables covalent binding to surfaces. (ii) C3a is an ANA that can bind to its receptor (C3aR), leading to pro- and antiinflammatory effects, and is expressed in most species. (iii) Once formed, C3b can interact with Factor B, properdin, and various complement regulators. Factor B binding initiates formation of C3-convertases, which cleaves additional C3 into C3b, thereby creating an amplification loop. Binding of FH, MCP, DAF, and CR1 mediates C3-convertases’ deactivation (via disruption of the C3b–Bb interaction) or degradation (via proteolytic cleavage of C3b). (iv) C3 contains two highly conserved cleavage sites for Factor I (FI), which, in the presence of cofactors such as FH, MCP, or CR1, inhibit further activation and cleave C3. The first cleavage by FI releases C3f, forming inactivated C3b (iC3b). A second cleavage releases C3c from the target-bound C3dg fragment. C3 fragments can still exert functional consequences via interaction with receptors.

Three examples of key paradigm shifts in the understanding of complement biology discovered in animal models and proven relevant to humans will be highlighted. Traditionally, the complement system was regarded as a liver-produced system confined to the circulation. However, research in mice revealed that locally produced complement is crucial for immune responses in diseases. Over 35 years ago, mouse kidneys were found to express and synthesize prominent amounts of complement (30). Later, a series of elegant experiments using a murine kidney transplantation model demonstrated that locally produced C3, rather than circulating C3, is paramount in initiating alloreactivity (31). When wild-type or C3–/– kidneys were transplanted into C3–/– or wild-type recipients, wild-type recipients of C3–/– kidneys exhibited the best outcomes, with 80% graft survival after 100 days. Recently, these observations were verified in humans, where genetic variations in donor C3, Factor B, and Factor H were associated with allograft survival in kidney transplantation, whereas recipient genetics had no effect (32). Another discovery arising from mice is the interaction between the LP and the alternative pathway (AP) via MASP-3, a splice variant of the MASP1 gene (33). Evidence of MASP-3’s role in AP activation came from MASP1/3–/– mice, which showed minimal AP activity alongside increased pro-enzyme Factor D levels (34, 35). MASP-1 was found to convert pro-Factor D in vitro (34, 35), but MASP-3 was ultimately uncovered as the main Factor D activator in vivo (36, 37). Findings in humans verified that MASP-3 functions similarly across mammals (38, 39). A final noteworthy example is sex-based differences in the complement system, first reported in Science in 1966 (40). Testosterone treatment in sterilized mice enhanced terminal complement activity, whereas estrogen decreased it (40). Recent work verified that female mice have reduced complement activity because of lower levels of terminal components (41). Complement assessments in healthy Norwegian blood donors corroborated that women have lower levels of terminal components and reduced functional activity (42).

Differences in the complement system among species. The complement system exhibits notable differences across humans and research animals (Tables 1–5). Even closely related primate species show divergences in complement genetics, circulating levels, and activity (43, 44). Among the pathways, the LP shows the greatest disparities across species (Table 1). In humans and great apes, MBL2 encodes mannose-binding lectin, while MBL1 is a pseudogene (45). In contrast, rodents, rabbits, pigs, and rhesus monkeys have two functional genes: Mbl-a and Mbl-c (45–48). Similarly, while humans and primates possess three ficolins (ficolin-1 to ficolin-3), rodents, rabbits, and pigs have only two (ficolin-A and ficolin-B), with ficolin-3 being a pseudogene (49–51). Additionally, mice exhibit reduced classical pathway (CP) functionality (52). C1r and C1s exist as gene duplicates in mice, whereas humans possess a single gene for each (53, 54). Humans have two C4-encoding genes (C4A and C4B), while mice have one C4 gene (Table 2), along with a “C4-like” gene for sex-limited protein (Slp) (55). Unlike C4, Slp is exclusively expressed in male mice of certain strains, is not cleaved by C1s, and has low C4 activity (56–58). However, Slp may enhance CP activation by acting synergistically with C4 (58). Furthermore, immunoglobulin (sub)classes differ across species in their ability to activate complement (59).

Functional assays indicate that the AP, compared with other pathways, is relatively more potent in rodents than in humans (60–62). Although Factor H shows high cross-species similarities (Table 3), this does not apply to other members of the Factor H protein family (63). Rodents lack an ortholog of human Factor H-like protein 1 (FHL-1), an alternative splicing variant of the Factor H gene (63). Humans also have Factor H-related proteins (FHR-1 to FHR-5), originating from duplication events of the Factor H gene, leading to structural similarities (64). However, these duplication events occurred after the divergence of rodent and primate lineages (64). Consequently, the structure, domain composition, and sequence of murine FHR genes differ from those in humans (63, 64). The resemblance between FHRs in humans and other animals, regarding distribution and functionality, remains unclear.

Surface regulators and receptors also exhibit major cross-species differences (Tables 4 and 5). Membrane cofactor protein (MCP/CD46) is widely expressed in humans but limited to testes in rodents (65). Furthermore, pigs, bovines, and most primates express MCP on erythrocytes, whereas humans do not (65–68). Although MCP’s cofactor activity for Factor I–mediated cleavage of C3b is conserved across species (66, 69), structural differences exist, as MCP is a receptor for species-specific pathogens (70). Decay-accelerating factor (DAF/CD55) and CD59 are other widely expressed surface regulators in pigs, primates, and humans, preventing complement-mediated cell lysis (71, 72). Mice, however, possess two genes for both regulators, one widely expressed and resembling its human counterpart, and one restricted to the testes (73–75). Remarkably, guinea pigs are the only mammals that lack CD59 (76).

In humans and primates, complement receptor 1 (CR1/CD35) and complement receptor 2 (CR2/CD21) are encoded by separate genes (77), while in rodents, a single gene (Cr2) produces both receptors via alternative splicing (78, 79). Additionally, key differences exist in structure, functionality, and expression between rodent and human CR1 and CR2 (77–84). Rodents express another regulator named CR1-related gene/protein Y (Crry), which is absent in humans and primates, likely performing regulatory roles of human DAF, MCP, and CR1 (83, 84). The loss of Crry in primates is believed to have contributed to the development of a separate CR1 gene (77). The dog genome contains a single Cr2-like gene adjacent to two Crry-like and two MCP-like genes, whereas the gene organization of complement receptors in pigs remains poorly characterized. Finally, although C3a and C5a receptors in humans and mice are functionally similar, their cellular expression shows both overlap and differences (Table 5) (85–87). Among GPCRs, which typically exhibit 85%–98% homology between humans and mice, anaphylatoxin receptors have the lowest homology (61%–65%) (88–90). Single-cell sequencing is enhancing cross-species comparisons of complement mRNA expression in cell types and tissues, revealing both similarities and differences (91, 92).

Species differences in the binding avidities of complement initiators, as well as the composition and potency of complement pathways, can cause divergences in the mechanism of complement activation between animal models and human diseases, despite both being complement mediated (93–95). Although differences in complement across species are often used to critique animal studies, they have also advanced our understanding of human diseases. Interspecies differences and animal studies have been pivotal in identifying MCP as the receptor for measles virus, which infects humans and primates but not rodents (70). MCP is highly homologous between humans and primates, while rodents exhibit key structural and expression differences (96, 97). Experiments with monkey erythrocytes first suggested MCP as a measles receptor. Functional studies with rodent cells provided conclusive evidence that the virus could bind to and infect rodent cells if they expressed human MCP but no native rodent MCP (96, 97). Thus, while differences in the complement system between animals and humans pose challenges for translational research, they can also help uncover human-specific biology.

Innovations in animal testing for complement research. Advances in genome engineering have enabled the development of animal models that more accurately mimic aspects of human physiology, enhancing their clinical relevance. Targeted genomic humanization and conditional or inducible gene knockouts in rodents and larger animals have improved biological alignment with humans.

Replacing animal genes with human equivalents has been employed to address interspecies differences and to aid preclinical drug testing of human-specific targets. Identifying human MCP as the measles virus receptor led to the creation of human MCP-transgenic mice, enabling measles infection studies in previously resistant animals (98). Furthermore, since rodents have a single gene (Cr2) for CR1 and CR2, Cr2–/– mice exhibit dual deficiencies. Mice expressing human complement receptors were therefore developed to study their individual roles in vivo (99–101). Similarly, since mice express a single C4 gene (C4b), introducing human C4A into mice helped uncover the mechanisms underlying the association between C4A and schizophrenia in humans (102, 103). However, transgenic expression of human complement has also had unexpected effects. Humanizing C3 in mice triggered C3 glomerulopathy (C3G), a complement-mediated kidney disease, because of impaired regulation of human C3 by mouse inhibitors, causing spontaneous complement activation (104). Alternatively, C3-humanized rats remain healthy and do not exhibit uncontrolled C3 activation (105). Furthermore, humanized Factor H mice normally regulate their AP and attenuate or reverse kidney and eye pathology seen in Cfh–/– mice (106). Other successful examples of transgenic rodents include knockins of human C1q, C5, C5aR1, C6, DAF, CD59, and C1 inhibitor (107–113).

The development of inducible and/or tissue-specific gene manipulation in mice enables spatial and temporal control in preclinical models. Early applications of this technology involved mice expressing human CD59 on erythrocytes or endothelial cells (114). Subsequent targeting of human CD59 in these mice with a pore-forming toxin created distinct disease models: disseminated intravascular coagulation when endothelial cells expressed CD59 and acute hemolysis when erythrocytes expressed CD59 (114). Tissue-specific knockout mice for properdin identified myeloid cells as the primary source of circulating properdin levels (115), while mice with a conditional deletion of Crry in proximal tubular epithelium circumvented the embryonic lethality seen in global knockout mice (116, 117). Overall, tissue- and cell-specific knockouts of complement genes have clarified the distinct functions of local versus systemic complement sources, and their relative significance in infections and inflammatory diseases, sometimes revealing opposing effects (118–121). These models have also uncovered key cell-intrinsic functions of complement (122, 123).

CRISPR/Cas technology has revolutionized genome editing, enabling multiple genetic modifications simultaneously. CRISPR/Cas has created mice with atypical hemolytic uremic syndrome–associated (aHUS-associated) mutations, verifying disease causality and facilitating preclinical drug testing (124, 125). CRISPR/Cas systems also allow for larger gene modifications, such as the humanization of the entire Factor H locus in mice (126). These mice lacked murine Factor H and FHRs but expressed human Factor H along with a normal or mutant FHR-5. The mutant FHR-5, linked to C3G in humans, resulted in a gain of function, causing C3 deposition in the kidney and spontaneous disease (126). Additionally, CRISPR/Cas has generated Serping1–/–, C1qa–/–, Masp3–/–, Cfd–/–, Cfhr-e–/–, and C5–/– mice (127–132). Traditional genetic modification methods were challenging for large animals; however, CRISPR has enabled the development of C3–/– pigs (133). Notably, CRISPR facilitated the creation of pigs with multiple genetic modifications, including human MCP and DAF expression, advancing xenotransplantation toward clinical application (134).

Evaluating complement-targeted therapies: animal models versus clinical trials

Some diseases have shown drug efficacy in human trials consistent with prior observations in animal models, yet in other cases, clinical trials have failed despite robust animal evidence. It is challenging to discern whether these failures stem from the limitations of animal models or flaws in trial design. Additionally, some anticomplement drugs have been approved based on successful clinical trials in diseases without extensive animal testing, suggesting that animal studies may not always be essential. Conversely, failed trials without strong evidence from animal models raise questions about whether animal studies could have improved the design or predicted failure. Last, although not discussed here, animal testing is crucial for assessing toxicity (discussed in ref. 135).

Animal studies leading to approved complement inhibitors. AAV is a group of diseases characterized by vascular inflammation in small vessels. This disease exemplifies how discoveries from animal models can lead to the clinical approval of novel treatments (7). Traditionally, AAV was not considered complement mediated, as circulating C3 and C4 levels are often normal, with minimal tissue deposits of immunoglobulin or complement (136). Clinical trials demonstrated that adding avacopan (a C5aR1 blocker) to existing immunosuppression regimens for maintaining disease remission facilitated faster glucocorticoid tapering, thereby reducing side effects, leading to FDA approval (6). Fifteen years prior, animal studies sparked interest in the complement system in AAV by identifying a critical role for the AP and showing that genetic deletion of Factor B and C5 provided protection (137). Mouse models further revealed the importance of the C5a/C5aR1 interaction in AAV pathophysiology (138). Finally, murine models demonstrated that anti-C5 therapy and blocking C5aR provide protection in AAV (139, 140). Analysis of patients with AAV shows AP activation fragments in blood, urine, and tissues (discussed in ref. 141), verifying findings from animal models. Although human studies may have eventually uncovered the involvement of complement, the initial findings in animal models profoundly accelerated this process.

aHUS is another disease where animal models contributed to and supported the effectiveness of clinical complement inhibition. AP involvement in aHUS was first reported in the 1970s (142–145). Genetic studies associated Factor H mutations, followed by other complement-related gene variants, with the disease (146–150). This suggested that complement activation is central to aHUS, though the precise mechanisms remained unclear. Factor H mutations had also been linked to C3G, which is diagnosed by prominent C3 deposition within the glomeruli (151). Animal models established a causal link between Factor H deficiencies and C3G, as a genetic deficiency in pigs led to spontaneous disease (152). Mice with targeted gene deletions of Factor H verified that complement dysregulation drives C3G (153). A key question remained: Why do some patients with complement dysregulation develop aHUS, while others develop C3G? Factor H mutations in aHUS clustered in the protein’s C-terminus, reducing protection of host cells from unwanted complement activation (154, 155). The seminal study by Pickering et al. revealed that complete Factor H deficiency led to C3G-like disease, whereas mice expressing a Factor H that lacked the last five domains developed aHUS-like disease (156). The structural similarities between Factor H in mice and humans enabled this breakthrough (25), offering the first in vivo evidence that impaired surface recognition by Factor H leads to aHUS. Of note, animal models showed that C5 inhibition in C3G provided only partial protection (157), foreshadowing mixed results in clinical trials with anti-C5 therapy (158). Recent studies suggest that aHUS and C3G probably require different therapeutic approaches to inhibit complement, as loss of Factor D or properdin in mouse models exacerbated C3G but protected against aHUS (125, 159–161).

Myasthenia gravis (MG) is an autoimmune disorder where autoantibodies disrupt neuromuscular transmission. Complement-activating autoantibodies are the primary driver of MG. In 1959, circulating levels of complement were already reported to inversely correlate with the severity of MG symptoms in patients (162). Fifteen years later, evidence emerged that targeting complement could treat MG, with C3 depletion being protective in a rat model (163). In MG patients and animal models, the MAC localizes at the neuromuscular junction (164–166). Animal studies conducted in the late 1980s predicted the success of terminal pathway inhibition in MG, as C5–/– mice were protected, while anti-C6 Fab antibodies in rats alleviated MG symptoms (167, 168). Overall, animal models have provided compelling evidence for the involvement of complement in MG and its therapeutic potential (169). Although a phase III study of anti-C5 therapy (eculizumab) in refractory MG missed its primary endpoint, positive secondary outcomes showed sustained benefit during the open-label extension phase, leading to FDA approval (170, 171). Later clinical trials with other anti-C5 therapies demonstrated greater improvements in generalized MG (172, 173), resulting in approval of ravulizumab (a long-acting monoclonal antibody against C5) and zilucoplan (a C5-blocking cyclic peptide).

Neuromyelitis optica spectrum disorder (NMOSD) is a relapsing inflammatory disease of the CNS, distinct from multiple sclerosis. Recently, pathogenic autoantibodies targeting the astrocytic water channel aquaporin 4 (AQP4) were identified in most patients with NMOSD, termed AQP4-IgG-seropositive NMOSD (174). In a randomized clinical trial involving patients with AQP4-IgG-seropositive NMOSD, eculizumab reduced the relative relapse risk by 94% compared with placebo (175). Later, ravulizumab demonstrated a similar reduction in relapse risk (176). Animal models of NMOSD were vital in establishing the pathogenic role of anti-AQP4 autoantibodies and the complement system (177–183). Mechanistically, these models uncovered that, in NMOSD, anti-AQP4 autoantibodies bind to astrocytes, triggering complement-mediated cell damage, leading to leukocyte infiltration, cytokine release, and blood-brain barrier disruption (184, 185). This ultimately causes bystander oligodendrocyte injury, myelin loss, and neuronal death (184, 185). Although no publications have reported on anti-C5 therapy in NMOSD models, complement knockouts and complement inhibitors validated the efficacy of complement-targeted therapies in NMOSD (186–188). Overall, the passive transfer of human anti-AQP4 autoantibodies in animal models has been instrumental for uncovering disease pathogenesis and identifying complement as a therapeutic target in NMOSD.

Unsuccessful clinical trials despite strong animal evidence. Outcomes in clinical trials of complement-targeted therapies have been most disappointing in ischemia-reperfusion injury (IRI) related to cardiovascular disease and transplantation, along with antibody-mediated transplant rejection. The first large clinical trials of anti-C5 therapy were conducted for cardiac IRI (189). Confidence in targeting complement arose from preclinical studies demonstrating its key role and the efficacy of inhibitors in reducing cardiac IRI in animal models (190, 191). Early studies of a single-chain antibody directed against C5 (pexelizumab) demonstrated promising results in patients with myocardial infarction undergoing reperfusion therapy (192–196). The Assessment of Pexelizumab in Acute Myocardial Infarction trial tested pexelizumab in 5,745 patients with acute myocardial infarction undergoing percutaneous coronary intervention to improve mortality (197). Additionally, the Pexelizumab for Reduction of Infarction and Mortality in Coronary Artery Bypass Graft Surgery–I and –II studies assessed pexelizumab in 3,099 and 4,254 patients receiving cardiac bypass surgery to reduce perioperative myocardial infarction and mortality (189, 198). Together, these trials did not show a consistent significant clinical improvement. Similarly, a soluble form of human CR1 (TP10) was tested in 564 high-risk cardiac surgery patients requiring cardiopulmonary bypass but failed to reduce morbidity or mortality, despite effectively inhibiting complement (199). Previous animal studies showed the inhibitor was highly effective in reducing cardiac IRI (200). Flaws in trial design have been suggested, and post hoc analyses proposed subgroups who might still benefit (201–205). However, it is crucial to recognize that animal models of cardiac IRI fail to accurately replicate the complex clinical setting of patients with myocardial infarction undergoing reperfusion therapy, including comorbidities and concomitant medications (206). For example, the use of heparin, which also affects complement activation, is a treatment aspect not replicated in animal models, potentially confounding results (207).

Although tested in smaller numbers of patients, clinical trials have extensively studied complement inhibitors in solid organ transplantation. Despite these efforts, no evident clinical improvements or regulatory approvals have been achieved to date (208). Clinical trials primarily focused on antibody-mediated rejection (AMR) and IRI and were conducted predominantly in kidney transplantation. Robust data from animal models, including rodents, pigs, and primates, consistently demonstrated the benefit of complement inhibition (209–213). Anti-C5 therapy with eculizumab has been evaluated in nine clinical trials for AMR in transplantation, for either prevention or treatment (ClinicalTrials.gov NCT02013037; NCT01399593; NCT01567085; NCT00670774; NCT01895127; NCT02113891; NCT01095887; NCT01106027; NCT01327573). Similarly, C1 inhibitor has been investigated in six clinical trials for AMR (ClinicalTrials.gov NCT01035593; NCT02936479; NCT02547220; NCT03221842; NCT01147302; NCT01134510). Blocking C5 did not significantly enhance outcomes in highly sensitized patients, nor did it prevent the progression to chronic AMR (214–219). Trials using C1 inhibitor in sensitized recipients also showed underwhelming results (220–222), but therapy may have been underdosed (223). Drugs targeting other complement proteins remain under active investigation in AMR (224). Additionally, complement-targeted drugs have been tested in clinical trials aimed at reducing IRI and improving short-term posttransplant outcomes (225–230), primarily in kidney transplantation. To date, there have been no concrete clinical advancements or regulatory approvals, despite various animal models of IRI predicting clinical success with anti-C5 therapy or C1 inhibitor (231–234).

Approved complement inhibitors without substantial animal studies. PNH was the first indication for which eculizumab received FDA approval (8, 9). It was well established that PNH erythrocytes lack CD55 and CD59 because of a genetic mutation affecting their glycosylphosphatidylinositol (GPI) anchor, leading to hemolysis via the insertion of C5b-9 (235). PNH exemplifies FDA approval of complement inhibitors with minimal animal studies. The reasons for limited animal studies in PNH are two-fold: (a) lack of representative animal models (236) and (b) ability to easily collect erythrocytes from affected patients (237), making it straightforward to study complement inhibitors in vitro. Moreover, prior clinical studies had demonstrated sufficient safety of C5 inhibition in humans. Subsequently, studies in patients with PNH treated with eculizumab also advanced our understanding of complement biology and disease mechanisms without further animal use, revealing C3 opsonins on erythrocytes trigger phagocytic uptake in the liver/spleen, causing extravascular hemolysis (10). Cold agglutinin disease (CAD) is another complement-mediated hemolytic anemia that received FDA approval for a complement inhibitor without comprehensive animal model testing. In CAD, autoreactive IgM activates the CP (238). However, since erythrocytes express CD55 and CD59, intravascular hemolysis by C5b-9 is limited and extravascular hemolysis predominates, driven by C3 opsonins (239). Like PNH, no accurate animal models exist, and anticomplement drug efficacy can be evaluated with in vitro assays using patient samples (240, 241). Sutimlimab, a C1s-blocking antibody, prevented opsonization of erythrocytes in vitro and was effective in a phase III study of patients with CAD, leading to FDA approval (241, 242). These results align with studies from the 1960s first suggesting complement’s role in extravascular hemolysis (243).

Age-related macular degeneration (AMD) is a multifactorial eye disease causing retinal degeneration and is the leading cause of blindness in the elderly population. Genetic studies in patients with AMD were the first to uncover the key role of the complement system (63). Nearly two decades ago, three independent studies identified a common genetic variant in Factor H that significantly increased disease risk (244–246). Later research uncovered variants in additional complement genes that contributed to disease risk (12, 247, 248), particularly in the FHRs (63). In 2023, the FDA approved two intravitreal complement inhibitors — pegcetacoplan (C3 inhibitor) and avacincaptad pegol (C5 inhibitor) — as the first treatments for advanced non-neovascular AMD (249–251). Although animal studies have supported and substantiated human genetic findings by confirming the causal role of complement in AMD (106, 252–258), their influence on the approval of complement inhibitors appears limited.

In IgA nephropathy (IgAN), clinical trials led to the approval of complement inhibitors without meaningful animal studies (259). Animal models of IgAN are limited and rarely used to test complement inhibition (260). Decades of observational human data strongly suggested an important role for complement in IgAN. In brief, kidney biopsy data demonstrated that glomerular complement deposition is nearly always present and holds prognostic value (summarized in ref. 261). Extensive biomarker evidence indicated AP activation, including tissue deposition and detection of activation fragments in plasma and urine (261). Unbiased genomics studies linked Factor H and FHR variants to disease risk and activity (13, 261), with circulating levels and renal deposits of FHR also associating with outcomes (63, 261). Collectively, these findings compellingly implicated the AP as a key driver in IgAN. An interim phase III trial analysis showed that iptacopan (Factor B inhibitor) significantly reduced proteinuria in patients with IgAN, leading to accelerated FDA approval (259). This success also highlights how clinical observations can provide a strong rationale for effective clinical trials. Ongoing follow-up will assess iptacopan’s impact on kidney function in IgAN.

A final example is CD55 deficiency with hyperactivation of complement, angiopathic thrombosis, and protein-losing enteropathy (CHAPLE) disease. In 2017, whole-exome sequencing associated loss-of-function variants in the DAF gene with early-onset protein-losing enteropathy and thrombosis in 11 individuals with gastrointestinal disorders, subsequently named CHAPLE disease (262). Shortly thereafter, reports demonstrated the efficacy of anti-C5 therapy for this condition (263). Pozelimab, a C5-blocking mAb, resolved clinical and laboratory manifestations of CHAPLE disease in 10 patients during an open-label phase II/III study (264), becoming the only FDA-approved treatment for this condition. Daf1–/– mice do not exhibit an evident phenotype but are more prone to complement-mediated inflammatory injury (265, 266). As in patients with CHAPLE, Daf1–/– mice exhibit heightened T cell activity and exacerbated autoimmune-induced colitis (267, 268). The swift approval of a complement inhibitor for CHAPLE disease clearly builds on insights from earlier translational and clinical studies of other complement-mediated diseases. However, given this existing knowledge and the availability of multiple clinical complement inhibitors with well-established safety and efficacy profiles, the necessity of additional animal studies for new indications could be questioned.

Discussion and remarks

We conclude that animal studies are not the only means of advancing disease understanding or developing complement-targeted therapies, as evidenced by the approval of complement therapeutics with and without reliance on animal studies. Simultaneously, we conclude that animal models remain a valuable tool in the complement field, which currently cannot be replaced. Increasingly available animal-free alternative research methods offer tools that supplement, rather than substitute for, animal-based approaches. Ultimately, every experiment must justify its choice of model — animal or otherwise — since all models are flawed and imperfect. Therapeutic targets supported by human observational genetic evidence are twice as likely to result in approved drugs than targets without human evidence (269). A recent meta-analysis estimated an 86% alignment in positive results between animal models and human studies for therapeutics, yet only 5% progress from animal studies to regulatory approval (270). This suggests that while animal models can accurately predict drug responses in human diseases, their translation is currently limited because of inconsistencies in design between preclinical studies and clinical trials. Therefore, aligning the design of animal studies with clinical trials — by incorporating randomization, blinding, clinically relevant outcomes, and long-term endpoints — could increase the number of treatments that progress from animal studies to regulatory approval. Currently, no data exist on the concordance between positive results from animal-free methods, e.g., organoids, and clinical trial outcomes. Until these methods are proven to be equally effective or superior, they cannot replace animal models.

The clinical efficacy of complement inhibitors is the ultimate validation of its pathophysiological relevance. However, the absence of clinical trials or negative clinical results does not necessarily disprove this. Industry chooses disease indications based on multiple factors, not just animal studies. While membranous nephropathy was among the first kidney diseases in which complement activation was thoroughly documented (271), industry prioritized clinical trials in IgAN. Furthermore, promising complement inhibitors in phase II trials have been discontinued because of shifting business priorities (272). Challenges such as patient recruitment for rare diseases, lengthy study durations, and complex endpoints in chronic conditions further complicate clinical trials. Design flaws may also contribute to unsuccessful outcomes (though this is speculative). For example, LP activation is observed in only one-third of patients with IgAN (261). However, a phase III trial of a MASP-2 inhibitor in IgAN proceeded without assessing LP activation and yielded negative results (NCT03608033). Furthermore, anti-C5 therapy was tested in membranous nephropathy but failed to reduce proteinuria (273). However, the study was prematurely halted, and concerns linger about the therapy being underdosed (274, 275), as proteinuria affects drug pharmacokinetics (276). In rheumatoid arthritis, complement inhibitors have yielded disappointing results so far, but they have been tested only in early-phase trials with few patients and short follow-up periods (277, 278). Consequently, the use of complement inhibitors in these diseases remains an unfinished story.

Animal models, when justified, are invaluable for exploring the complement system in health and disease. We believe this is also evident from our better understanding of disease mechanisms in conditions with approved complement inhibitors, such as aHUS, AAV, and MG, which have animal models, compared with CHAPLE and IgAN, which do not. Additionally, there is a clear need for better diagnostic tools for complement therapeutics, and animal models are extremely useful for developing and validating these tools, such as imaging approaches to detect tissue-bound complement deposits (279). Finally, animal studies have revealed surprising insights into complement’s role in disease, such as the discovery that complement activation can promote tumor growth in animal models (280, 281). While the translation to human disease is tentative, ongoing clinical trials of complement inhibitors for cancer will hopefully answer this question (NCT04919629; NCT04812535). Nevertheless, this research has already expanded our understanding of complement biology (1). In conclusion, when appropriately justified — particularly in relation to translation to human biology and disease — while always considering and addressing ethical concerns, animal models remain a valuable ally to the complement field in the foreseeable future, as they cannot yet be fully replaced.

Supplemental material

View Supplemental table 1

Acknowledgments

FP was supported by a K99 career development grant from the NIH National Institute of Diabetes and Digestive and Kidney Diseases (1K99DK138301-01A1). FP was also supported by a grant from the American Society of Nephrology Foundation for Kidney Research Ben J. Lipps Research Fellowship Program. Support also came from NIH R01DK076690 (to JMT) and R01DK138960 (to JMT and VMH).

Address correspondence to: Felix Poppelaars, Division of Nephrology and Hypertension, University of Colorado School of Medicine, B-115 1775 Aurora Court, M20-3103, Aurora, Colorado 80045, USA. Email: felix.poppelaars@cuanschutz.edu. Or to: Joshua M. Thurman, Division of Nephrology and Hypertension, University of Colorado School of Medicine, B-115 1775 Aurora Court, M20-3103, Aurora, Colorado 80045, USA. Email: joshua.thurman@cuanschutz.edu.

Footnotes

Conflict of interest: FP owns or owned stock in Apellis Pharmaceuticals, Annexon Biosciences, Chemocentryx, InflaRx, Iveric Bio, as well as Omeros Corporation and has been involved as a consultant for Alnylam Pharmaceuticals. JMT and VMH are consultants for Q32 Bio, Inc., a company developing complement inhibitors. Both also hold stock and may receive royalty income from Q32 Bio, Inc.

Copyright: © 2025, Poppelaars et al. This is an open access article published under the terms of the Creative Commons Attribution 4.0 International License.

Reference information: J Clin Invest. 2025;135(12):e188347. https://doi.org/10.1172/JCI188347.

References
  1. Mastellos DC, et al. A guide to complement biology, pathology and therapeutic opportunity. Nat Rev Immunol. 2024;24(2):118–141.
    View this article via: CrossRef PubMed Google Scholar
  2. Nonaka M, Kimura A. Genomic View of the Evolution of the Complement System. Springer; 2006.
  3. Holers VM. Contributions of animal models to mechanistic understandings of antibody-dependent disease and roles of the amplification loop. Immunol Rev. 2023;313(1):181–193.
    View this article via: CrossRef PubMed Google Scholar
  4. West EE, et al. Complement in human disease: approved and up-and-coming therapeutics. Lancet. 2024;403(10424):392–405.
    View this article via: CrossRef PubMed Google Scholar
  5. Yan B, et al. SARS-CoV-2 drives JAK1/2-dependent local complement hyperactivation. Sci Immunol. 2021;6(58):eabg0833.
    View this article via: CrossRef PubMed Google Scholar
  6. Jayne DRW, et al. Avacopan for the treatment of ANCA-associated vasculitis. N Engl J Med. 2021;384(7):599–609.
    View this article via: CrossRef PubMed Google Scholar
  7. Kallenberg CGM, Heeringa P. Complement system activation in ANCA vasculitis: a translational success story? Mol Immunol. 2015;68(1):53–56.
    View this article via: CrossRef PubMed Google Scholar
  8. Hillmen P, et al. Effect of eculizumab on hemolysis and transfusion requirements in patients with paroxysmal nocturnal hemoglobinuria. N Engl J Med. 2004;350(6):552–559.
    View this article via: CrossRef PubMed Google Scholar
  9. Hillmen P, et al. The complement inhibitor eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med. 2006;355(12):1233–1243.
    View this article via: CrossRef PubMed Google Scholar
  10. Schmidt CQ, et al. The complement model disease paroxysmal nocturnal hemoglobinuria. Eur J Immunol. 2024;54(11):e2350817.
    View this article via: CrossRef PubMed Google Scholar
  11. Kiryluk K, et al. Discovery of new risk loci for IgA nephropathy implicates genes involved in immunity against intestinal pathogens. Nat Genet. 2014;46(11):1187–1196.
    View this article via: CrossRef PubMed Google Scholar
  12. Hughes AE, et al. A common CFH haplotype, with deletion of CFHR1 and CFHR3, is associated with lower risk of age-related macular degeneration. Nat Genet. 2006;38(10):1173–1177.
    View this article via: CrossRef PubMed Google Scholar
  13. Kiryluk K, et al. Genome-wide association analyses define pathogenic signaling pathways and prioritize drug targets for IgA nephropathy. Nat Genet. 2023;55(7):1091–1105.
    View this article via: CrossRef PubMed Google Scholar
  14. Cervia-Hasler C, et al. Persistent complement dysregulation with signs of thromboinflammation in active Long Covid. Science. 2024;383(6680):eadg7942.
    View this article via: CrossRef PubMed Google Scholar
  15. Gaykema LH, et al. Inhibition of complement activation by CD55 overexpression in human induced pluripotent stem cell derived kidney organoids. Front Immunol. 2023;13:1058763.
    View this article via: CrossRef PubMed Google Scholar
  16. Miguel-Batuecas A, et al. Animal research in spain: a study of public perception and attitudes. Animals. 2023;13(12):2039.
    View this article via: CrossRef PubMed Google Scholar
  17. Rice M, et al. The impact of a negative media event on public attitudes towards animal welfare in the red meat industry. Animals. 2020;10(4):619.
    View this article via: CrossRef PubMed Google Scholar
  18. Metzger MM. Knowledge of the animal welfare act and animal welfare regulations influences attitudes toward animal research. J Am Assoc Lab Anim Sci. 2015;54(1):70–75.
    View this article via: PubMed Google Scholar
  19. Louis-Maerten E, et al. Conceptual foundations for a clarified meaning of the 3Rs principles in animal experimentation. Anim Welf. 2024;33:e37.
    View this article via: CrossRef PubMed Google Scholar
  20. Nonaka M. Evolution of the complement system. Subcell Biochem. 2014;80:31–43.
    View this article via: CrossRef PubMed Google Scholar
  21. Gibson BG, et al. Contribution of animal models to the mechanistic understanding of Alternative Pathway and Amplification Loop (AP/AL)-driven Complement-mediated Diseases. Immunol Rev. 2023;313(1):194–216.
    View this article via: CrossRef PubMed Google Scholar
  22. Ricklin D, et al. Complement component C3 - The “Swiss Army Knife” of innate immunity and host defense. Immunol Rev. 2016;274(1):33–58.
    View this article via: CrossRef PubMed Google Scholar
  23. Janssen BJC, et al. Structures of complement component C3 provide insights into the function and evolution of immunity. Nature. 2005;437(7058):505–511.
    View this article via: CrossRef PubMed Google Scholar
  24. Selman L, Hansen S. Structure and function of collectin liver 1 (CL-L1) and collectin 11 (CL-11, CL-K1). Immunobiology. 2012;217(9):851–863.
    View this article via: CrossRef PubMed Google Scholar
  25. Cheng ZZ, et al. Comparison of surface recognition and C3b binding properties of mouse and human complement factor H. Mol Immunol. 2006;43(7):972–979.
    View this article via: CrossRef PubMed Google Scholar
  26. Fakhouri F, et al. Treatment with human complement factor H rapidly reverses renal complement deposition in factor H-deficient mice. Kidney Int. 2010;78(3):279–286.
    View this article via: CrossRef PubMed Google Scholar
  27. Duncan AR, Winter G. The binding site for C1q on IgG. Nature. 1988;332(6166):738–740.
    View this article via: CrossRef PubMed Google Scholar
  28. Nonaka M, Miyazawa S. Evolution of the initiating enzymes of the complement system. Genome Biol. 2002;3(1):REVIEWS1001.
    View this article via: PubMed CrossRef Google Scholar
  29. Couves EC, et al. Structural basis for membrane attack complex inhibition by CD59. Nat Commun. 2023;14(1):890.
    View this article via: CrossRef PubMed Google Scholar
  30. Passwell J, et al. Local extrahepatic expression of complement genes C3, factor B, C2, and C4 is increased in murine lupus nephritis. J Clin Invest. 1988;82(5):1676–1684.
    View this article via: JCI CrossRef PubMed Google Scholar
  31. Pratt JR, et al. Local synthesis of complement component C3 regulates acute renal transplant rejection. Nat Med. 2002;8(6):582–587.
    View this article via: CrossRef PubMed Google Scholar
  32. Poppelaars F, et al. Synergistic impact of three complement polymorphisms in the donor, not the recipient, on long-term kidney allograft survival [preprint]. https://doi.org/10.1101/2023.10.24.23297481 Posted on medRxiv October 25, 2023.
  33. Dahl MR, et al. MASP-3 and its association with distinct complexes of the mannan-binding lectin complement activation pathway. Immunity. 2001;15(1):127–135.
    View this article via: CrossRef PubMed Google Scholar
  34. Takahashi M, et al. Essential role of mannose-binding lectin-associated serine protease-1 in activation of the complement factor D. J Exp Med. 2010;207(1):29–37.
    View this article via: CrossRef PubMed Google Scholar
  35. Takahashi M, et al. Mannose-binding lectin (MBL)-associated serine protease (MASP)-1 contributes to activation of the lectin complement pathway. J Immunol. 2008;180(9):6132–6138.
    View this article via: CrossRef PubMed Google Scholar
  36. Oroszlán G, et al. MASP-1 and MASP-2 do not activate pro-factor D in resting human blood, whereas MASP-3 is a potential activator: kinetic analysis involving specific MASP-1 and MASP-2 inhibitors. J Immunol. 2016;196(2):857–865.
    View this article via: CrossRef PubMed Google Scholar
  37. Dobó J, et al. MASP-3 is the exclusive pro-factor D activator in resting blood: the lectin and the alternative complement pathways are fundamentally linked. Sci Rep. 2016;6(1):31877.
    View this article via: CrossRef PubMed Google Scholar
  38. Hayashi M, et al. Cutting edge: role of MASP-3 in the physiological activation of factor D of the alternative complement pathway. J Immunol. 2019;203(6):1411–1416.
    View this article via: CrossRef PubMed Google Scholar
  39. Pihl R, et al. Analysis of factor D isoforms in malpuech-michels-mingarelli-carnevale patients highlights the role of MASP-3 as a maturase in the alternative pathway of complement. J Immunol. 2017;199(6):2158–2170.
    View this article via: CrossRef PubMed Google Scholar
  40. Weintraub RM, et al. Mouse complement: influence of sex hormones on its activity. Science. 1966;152(3723):783–785.
    View this article via: CrossRef PubMed Google Scholar
  41. Kotimaa J, et al. Sex matters: Systemic complement activity of female C57BL/6J and BALB/cJ mice is limited by serum terminal pathway components. Mol Immunol. 2015;76:13–21.
    View this article via: CrossRef PubMed Google Scholar
  42. Gaya da Costa M, et al. Age and sex-associated changes of complement activity and complement levels in a healthy caucasian population. Front Immunol. 2018;9:2664.
    View this article via: CrossRef PubMed Google Scholar
  43. Schur PH, et al. Phylogeny of complement components in non-human primates. J Immunol. 1975;114(1 pt 2):270–273.
    View this article via: CrossRef PubMed Google Scholar
  44. Xu H, et al. Investigation of cynomolgus monkey complement. Transplant Proc. 2008;40(2):607–608.
    View this article via: CrossRef PubMed Google Scholar
  45. Garred P, et al. A journey through the lectin pathway of complement-MBL and beyond. Immunol Rev. 2016;274(1):74–97.
    View this article via: CrossRef PubMed Google Scholar
  46. Genster N, et al. Lessons learned from mice deficient in lectin complement pathway molecules. Mol Immunol. 2014;61(2):59–68.
    View this article via: CrossRef PubMed Google Scholar
  47. Verga Falzacappa MV, et al. Evolution of the mannose-binding lectin gene in primates. Genes Immun. 2004;5(8):653–661.
    View this article via: CrossRef PubMed Google Scholar
  48. Juul-Madsen HR, et al. Structural gene variants in the porcine mannose-binding lectin 1 (MBL1) gene are associated with low serum MBL-A concentrations. Immunogenetics. 2011;63(5):309–317.
    View this article via: CrossRef PubMed Google Scholar
  49. Ohashi T, Erickson HP. Oligomeric structure and tissue distribution of ficolins from mouse, pig and human. Arch Biochem Biophys. 1998;360(2):223–232.
    View this article via: CrossRef PubMed Google Scholar
  50. Garred P, et al. The genetics of ficolins. J Innate Immun. 2010;2(1):3–16.
    View this article via: CrossRef PubMed Google Scholar
  51. Hummelshøj T, et al. Allelic lineages of the ficolin genes (FCNs) are passed from ancestral to descendant primates. PLoS One. 2011;6(12):e28187.
    View this article via: CrossRef PubMed Google Scholar
  52. Ratelade J, Verkman AS. Inhibitor(s) of the classical complement pathway in mouse serum limit the utility of mice as experimental models of neuromyelitis optica. Mol Immunol. 2014;62(1):104–113.
    View this article via: CrossRef PubMed Google Scholar
  53. Garnier G, et al. Complement C1r and C1s genes are duplicated in the mouse: differential expression generates alternative isomorphs in the liver and in the male reproductive system. Biochem J. 2003;371(pt 2):631–640.
    View this article via: CrossRef PubMed Google Scholar
  54. Arlaud GJ, et al. Structure, function and molecular genetics of human and murine C1r. Immunobiology. 2002;205(4–5):365–382.
    View this article via: CrossRef PubMed Google Scholar
  55. Blanchong CA, et al. Genetic, structural and functional diversities of human complement components C4A and C4B and their mouse homologues, Slp and C4. Int Immunopharmacol. 2001;(1):365–392.
    View this article via: CrossRef PubMed Google Scholar
  56. Ogata RT, et al. Substrate specificities of murine C1s. J Immunol. 1994;152(12):5890–5895.
    View this article via: CrossRef PubMed Google Scholar
  57. Nonaka M, et al. Complete nucleotide and derived amino acid sequences of sex-limited protein (Slp), nonfunctional isotype of the fourth component of mouse complement (C4). J Immunol. 1986;136(8):2989–2993.
    View this article via: CrossRef PubMed Google Scholar
  58. Beurskens FJM, et al. Mouse complement components C4 and Slp act synergistically in a homologous hemolytic C4 assay. Eur J Immunol. 2000;30(5):1507–1511.
    View this article via: CrossRef PubMed Google Scholar
  59. Mestas J, Hughes CCW. Of mice and not men: differences between mouse and human immunology. J Immunol. 2004;172(5):2731–2738.
    View this article via: CrossRef PubMed Google Scholar
  60. Lachmann PJ. Preparing serum for functional complement assays. J Immunol Methods. 2010;352(1–2):195–197.
    View this article via: CrossRef PubMed Google Scholar
  61. Ong GL, Mattes MJ. Mouse strains with typical mammalian levels of complement activity. J Immunol Methods. 1989;125(1–2):147–158.
    View this article via: CrossRef PubMed Google Scholar
  62. Kotimaa J. Analysis of Systemic Complement in Experimental Renal Injury and Disease. Doctoral Thesis. Leiden University; 2017.
  63. Poppelaars F, et al. A family affair: addressing the challenges of factor H and the related proteins. Front Immunol. 2021;12:660194.
    View this article via: CrossRef PubMed Google Scholar
  64. Cantsilieris S, et al. Recurrent structural variation, clustered sites of selection, and disease risk for the complement factor H (CFH) gene family. Proc Natl Acad Sci U S A. 2018;115(19):E4433–E4442.
    View this article via: CrossRef PubMed Google Scholar
  65. Tsujimura A, et al. Molecular cloning of a murine homologue of membrane cofactor protein (CD46): preferential expression in testicular germ cells. Biochem J. 1998;330(pt 1):163–168.
    View this article via: CrossRef PubMed Google Scholar
  66. van den Berg CW, et al. Purification and characterization of the pig analogue of human membrane cofactor protein (CD46/MCP). J Immunol. 1997;158(4):1703–1709.
    View this article via: CrossRef PubMed Google Scholar
  67. Antalíková J, et al. Identification of MCP/CD46 analogue on bovine erythrocytes using the new monoclonal antibody IVA-520. Vet Immunol Immunopathol. 2007;115(1–2):155–159.
    View this article via: CrossRef PubMed Google Scholar
  68. Nickells MW, Atkinson JP. Characterization of CR1- and membrane cofactor protein-like proteins of two primates. J Immunol. 1990;144(11):4262–4268.
    View this article via: CrossRef PubMed Google Scholar
  69. Seya T, et al. CD46 (membrane cofactor protein of complement, measles virus receptor): structural and functional divergence among species (review). Int J Mol Med. 1998;1(5):809–816.
    View this article via: PubMed CrossRef Google Scholar
  70. Dörig RE, et al. CD46, a primate-specific receptor for measles virus. Trends Microbiol. 1994;2(9):312–318.
    View this article via: CrossRef PubMed Google Scholar
  71. Miwa T, Song WC. Membrane complement regulatory proteins: Insight from animal studies and relevance to human diseases. Int Immunopharmacol. 2001;(3):445–459.
    View this article via: CrossRef PubMed Google Scholar
  72. Hinchliffe SJ, et al. Molecular cloning and functional characterization of the pig analogue of CD59: relevance to xenotransplantation. J Immunol. 1998;160(8):3924–3932.
    View this article via: CrossRef PubMed Google Scholar
  73. Spicer AP, et al. Molecular cloning and chromosomal localization of the mouse decay-accelerating factor genes. Duplicated genes encode glycosylphosphatidylinositol-anchored and transmembrane forms. J Immunol. 1995;155(6):3079–3091.
    View this article via: CrossRef PubMed Google Scholar
  74. Powell MB, et al. Molecular cloning, chromosomal localization, expression, and functional characterization of the mouse analogue of human CD59. J Immunol. 1997;158(4):1692–1702.
    View this article via: CrossRef PubMed Google Scholar
  75. Harris CL, et al. Characterization of the mouse analogues of CD59 using novel monoclonal antibodies: tissue distribution and functional comparison. Immunology. 2003;109(1):117–126.
    View this article via: CrossRef PubMed Google Scholar
  76. Boshra H, et al. Absence of CD59 in Guinea pigs: analysis of the cavia porcellus genome suggests the evolution of a CD59 pseudogene. J Immunol. 2018;200(1):327–335.
    View this article via: CrossRef PubMed Google Scholar
  77. Birmingham DJ, Hebert LA. CR1 and CR1-like: the primate immune adherence receptors. Immunol Rev. 2001;180:100–111.
    View this article via: CrossRef PubMed Google Scholar
  78. Jacobson AC, Weis JH. Comparative functional evolution of human and mouse CR1 and CR2. J Immunol. 2008;181(5):2953–2959.
    View this article via: CrossRef PubMed Google Scholar
  79. Michael Holers V, et al. The evolution of mouse and human complement C3-binding proteins: divergence of form but conservation of function. Immunol Today. 1992;13(6):231–236.
    View this article via: CrossRef PubMed Google Scholar
  80. Cosio FG, et al. Evaluation of the mechanisms responsible for the reduction in erythrocyte complement receptors when immune complexes form in vivo in primates. J Immunol. 1990;145(12):4198–4206.
    View this article via: CrossRef PubMed Google Scholar
  81. Alexander JJ, et al. A protein with characteristics of factor H is present on rodent platelets and functions as the immune adherence receptor. J Biol Chem. 2001;276(34):32129–32135.
    View this article via: CrossRef PubMed Google Scholar
  82. Alexander JJ, et al. Mouse podocyte complement factor H: the functional analog to human complement receptor 1. J Am Soc Nephrol. 2007;18(4):1157–1166.
    View this article via: CrossRef PubMed Google Scholar
  83. Kim YU, et al. Mouse complement regulatory protein Crry/p65 uses the specific mechanisms of both human decay-accelerating factor and membrane cofactor protein. J Exp Med. 1995;181(1):151–159.
    View this article via: CrossRef PubMed Google Scholar
  84. Molina H, et al. Distinct receptor and regulatory properties of recombinant mouse complement receptor 1 (CR1) and Crry, the two genetic homologues of human CR1. J Exp Med. 1992;175(1):121–129.
    View this article via: CrossRef PubMed Google Scholar
  85. Yadav MK, et al. Molecular basis of anaphylatoxin binding, activation, and signaling bias at complement receptors. Cell. 2023;186(22):4956–4973.
    View this article via: CrossRef PubMed Google Scholar
  86. Gorman DM, et al. Development of synthetic human and mouse C5a: application to binding and functional assays in vitro and in vivo. ACS Pharmacol Transl Sci. 2021;4(6):1808–1817.
    View this article via: CrossRef PubMed Google Scholar
  87. Laumonnier Y, et al. Novel insights into the expression pattern of anaphylatoxin receptors in mice and men. Mol Immunol. 2017;89:44–58.
    View this article via: CrossRef PubMed Google Scholar
  88. Bamberg CE, et al. The C5a receptor (C5aR) C5L2 is a modulator of C5aR-mediated signal transduction. J Biol Chem. 2010;285(10):7633–7644.
    View this article via: CrossRef PubMed Google Scholar
  89. Monk PN, et al. Function, structure and therapeutic potential of complement C5a receptors. Br J Pharmacol. 2007;152(4):429–448.
    View this article via: CrossRef PubMed Google Scholar
  90. Gerard C, et al. Structural diversity in the extracellular faces of peptidergic G-protein-coupled receptors. Molecular cloning of the mouse C5a anaphylatoxin receptor. J Immunol. 1992;149(8):2600–2606.
    View this article via: CrossRef PubMed Google Scholar
  91. Chaudhary N, et al. A single-cell lung atlas of complement genes identifies the mesothelium and epithelium as prominent sources of extrahepatic complement proteins. Mucosal Immunol. 2022;15(5):927–939.
    View this article via: CrossRef PubMed Google Scholar
  92. Zilionis R, et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity. 2019;50(5):1317–1334.
    View this article via: CrossRef PubMed Google Scholar
  93. Tirado TC, et al. A comparative approach on the activation of the three complement system pathways in different hosts of Visceral Leishmaniasis after stimulation with Leishmania infantum. Dev Comp Immunol. 2021;120:104061.
    View this article via: CrossRef PubMed Google Scholar
  94. Li Y, et al. Complement opsonization of nanoparticles: Differences between humans and preclinical species. J Control Release. 2021;338:548–556.
    View this article via: CrossRef PubMed Google Scholar
  95. van deer Pol P, et al. Natural IgM antibodies are involved in the activation of complement by hypoxic human tubular cells. Am J Physiol Renal Physiol. 2011;300(4):F932–F940.
    View this article via: CrossRef PubMed Google Scholar
  96. Dörig RE, et al. The human CD46 molecule is a receptor for measles virus (Edmonston strain). Cell. 1993;75(2):295–305.
    View this article via: CrossRef PubMed Google Scholar
  97. Naniche D, et al. Human membrane cofactor protein (CD46) acts as a cellular receptor for measles virus. J Virol. 1993;67(10):6025–6032.
    View this article via: CrossRef PubMed Google Scholar
  98. Rall GF, et al. A transgenic mouse model for measles virus infection of the brain. Proc Natl Acad Sci U S A. 1997;94(9):4659–4663.
    View this article via: CrossRef PubMed Google Scholar
  99. Marchbank KJ, et al. Expression of human complement receptor 2 (CR2, CD21) in Cr2-/- mice restores humoral immune function. J Immunol. 2000;165(5):2354–2361.
    View this article via: CrossRef PubMed Google Scholar
  100. Repik A, et al. A transgenic mouse model for studying the clearance of blood-borne pathogens via human complement receptor 1 (CR1). Clin Exp Immunol. 2005;140(2):230–240.
    View this article via: CrossRef PubMed Google Scholar
  101. Jackson HM, et al. A novel mouse model expressing human forms for complement receptors CR1 and CR2. BMC Genet. 2020;21(1):101.
    View this article via: CrossRef PubMed Google Scholar
  102. Yilmaz M, et al. Overexpression of schizophrenia susceptibility factor human complement C4A promotes excessive synaptic loss and behavioral changes in mice. Nat Neurosci. 2021;24(2):214–224.
    View this article via: CrossRef PubMed Google Scholar
  103. Sekar A, et al. Schizophrenia risk from complex variation of complement component 4. Nature. 2016;530(7589):177–183.
    View this article via: CrossRef PubMed Google Scholar
  104. Devalaraja-Narashimha K, et al. Humanized C3 mouse: a novel accelerated model of C3 glomerulopathy. J Am Soc Nephrol. 2021;32(1):99–114.
    View this article via: CrossRef PubMed Google Scholar
  105. Chen JY, et al. Development of a C3 humanized rat as a new model for evaluating novel C3 inhibitors. J Innate Immun. 2023;16(1):56–65.
    View this article via: CrossRef PubMed Google Scholar
  106. Ding JD, et al. Expression of human complement factor H prevents age-related macular degeneration-like retina damage and kidney abnormalities in aged Cfh knockout mice. Am J Pathol. 2015;185(1):29–42.
    View this article via: CrossRef PubMed Google Scholar
  107. Li M, et al. Development of a humanized C1q A chain knock-in mouse: assessment of antibody independent beta-amyloid induced complement activation. Mol Immunol. 2008;45(11):3244–3252.
    View this article via: CrossRef PubMed Google Scholar
  108. Latuszek A, et al. Inhibition of complement pathway activation with Pozelimab, a fully human antibody to complement component C5. PLoS One. 2020;15(5):e0231892.
    View this article via: CrossRef PubMed Google Scholar
  109. Lee H, et al. Human C5aR knock-in mice facilitate the production and assessment of anti-inflammatory monoclonal antibodies. Nat Biotechnol. 2006;24(10):1279–1284.
    View this article via: CrossRef PubMed Google Scholar
  110. Gytz Olesen H, et al. Development, characterization, and in vivo validation of a humanized C6 monoclonal antibody that inhibits the membrane attack complex. J Innate Immun. 2023;15(1):16–36.
    View this article via: CrossRef PubMed Google Scholar
  111. Van Denderen BJW, et al. Expression of functional decay-accelerating factor (CD55) in transgenic mice protects against human complement-mediated attack. Transplantation. 1996;61(4):582–588.
    View this article via: CrossRef PubMed Google Scholar
  112. Diamond LE, et al. Human CD59 expressed in transgenic mouse hearts inhibits the activation of complement. Transpl Immunol. 1995;3(4):305–312.
    View this article via: CrossRef PubMed Google Scholar
  113. Vinci G, et al. In vivo biosynthesis of endogenous and of human C1 inhibitor in transgenic mice: tissue distribution and colocalization of their expression. J Immunol. 2002;169(10):5948–5954.
    View this article via: CrossRef PubMed Google Scholar
  114. Hu W, et al. Rapid conditional targeted ablation of cells expressing human CD59 in transgenic mice by intermedilysin. Nat Med. 2008;14(1):98–103.
    View this article via: CrossRef PubMed Google Scholar
  115. Kimura Y, et al. Genetic and therapeutic targeting of properdin in mice prevents complement-mediated tissue injury. J Clin Invest. 2010;120(10):3545–3554.
    View this article via: JCI CrossRef PubMed Google Scholar
  116. Miao J, et al. Tissue-specific deletion of Crry from mouse proximal tubular epithelial cells increases susceptibility to renal ischemia-reperfusion injury. Kidney Int. 2014;86(4):726–737.
    View this article via: CrossRef PubMed Google Scholar
  117. Xu C, et al. A critical role for murine complement regulator crry in fetomaternal tolerance. Science. 2000;287(5452):498–501.
    View this article via: CrossRef PubMed Google Scholar
  118. Sahu SK, et al. Lung epithelial cell-derived C3 protects against pneumonia-induced lung injury. Sci Immunol. 2023;8(80):eabp9547.
    View this article via: CrossRef PubMed Google Scholar
  119. Desai JV, et al. C5a-licensed phagocytes drive sterilizing immunity during systemic fungal infection. Cell. 2023;186(13):2802–2822.
    View this article via: CrossRef PubMed Google Scholar
  120. Yu Q, et al. C1q is essential for myelination in the central nervous system (CNS). iScience. 2023;26(12):108518.
    View this article via: CrossRef PubMed Google Scholar
  121. Wu M, et al. Gut complement induced by the microbiota combats pathogens and spares commensals. Cell. 2024;187(4):897–913.
    View this article via: CrossRef PubMed Google Scholar
  122. West EE, Kemper C. Complosome - the intracellular complement system. Nat Rev Nephrol. 2023;19(7):426–439.
    View this article via: CrossRef PubMed Google Scholar
  123. King BC, Blom AM. Intracellular complement and immunometabolism: The advantages of compartmentalization. Eur J Immunol. 2024;54(8):e2350813.
    View this article via: CrossRef PubMed Google Scholar
  124. Smith-Jackson K, et al. Hyperfunctional complement C3 promotes C5-dependent atypical hemolytic uremic syndrome in mice. J Clin Invest. 2019;129(3):1061–1075.
    View this article via: JCI CrossRef PubMed Google Scholar
  125. Ueda Y, et al. Murine systemic thrombophilia and hemolytic uremic syndrome from a factor H point mutation. Blood. 2017;129(9):1184–1196.
    View this article via: CrossRef PubMed Google Scholar
  126. Malik TH, et al. Gain-of-function factor H–related 5 protein impairs glomerular complement regulation resulting in kidney damage. Proc Natl Acad Sci U S A. 2021;118(13):e2022722118.
    View this article via: CrossRef PubMed Google Scholar
  127. Tsuru H, et al. HFD-induced hepatic lipid accumulation and inflammation are decreased in Factor D deficient mouse. Sci Rep. 2020;10(1):17593.
    View this article via: CrossRef PubMed Google Scholar
  128. Li X, et al. Deficiency of mouse FHR-1 homolog, FHR-E, accelerates sepsis, and acute kidney injury through enhancing the LPS-induced alternative complement pathway. Front Immunol. 2020;11:1123.
    View this article via: CrossRef PubMed Google Scholar
  129. Qiu T, et al. Gene therapy for C1 esterase inhibitor deficiency in a Murine Model of Hereditary angioedema. Allergy. 2019;74(6):1081–1089.
    View this article via: CrossRef PubMed Google Scholar
  130. Gullipalli D, et al. MASP3 deficiency in mice reduces but does not abrogate alternative pathway complement activity due to intrinsic profactor D activity. J Immunol. 2023;210(10):1543–1551.
    View this article via: CrossRef PubMed Google Scholar
  131. Winn NC, et al. Deletion of complement factor 5 amplifies glucose intolerance in obese male but not female mice. Am J Physiol Endocrinol Metab. 2023;325(4):E325–E335.
    View this article via: CrossRef PubMed Google Scholar
  132. Kim HW, et al. C1qa deficiency in mice increases susceptibility to mouse hepatitis virus A59 infection. J Vet Sci. 2021;22(3):e36.
    View this article via: CrossRef PubMed Google Scholar
  133. Zhang W, et al. Generation of complement protein C3 deficient pigs by CRISPR/Cas9-mediated gene targeting. Sci Rep. 2017;7(1):5009.
    View this article via: CrossRef PubMed Google Scholar
  134. Anand RP, et al. Design and testing of a humanized porcine donor for xenotransplantation. Nature. 2023;622(7982):393–401.
    View this article via: CrossRef PubMed Google Scholar
  135. Prior H, et al. Exploring greater flexibility for chronic toxicity study designs to support human safety assessment while balancing 3Rs considerations. Int J Toxicol. 2024;43(5):456–463.
    View this article via: CrossRef PubMed Google Scholar
  136. Hebert LA, et al. Diagnostic significance of hypocomplementemia. Kidney Int. 1991;39(5):811–821.
    View this article via: CrossRef PubMed Google Scholar
  137. Xiao H, et al. Alternative complement pathway in the pathogenesis of disease mediated by anti-neutrophil cytoplasmic autoantibodies. Am J Pathol. 2007;170(1):52–64.
    View this article via: CrossRef PubMed Google Scholar
  138. Schreiber A, et al. C5a receptor mediates neutrophil activation and ANCA-induced glomerulonephritis. J Am Soc Nephrol. 2009;20(2):289–298.
    View this article via: CrossRef PubMed Google Scholar
  139. Xiao H, et al. C5a receptor (CD88) blockade protects against MPO-ANCA GN. J Am Soc Nephrol. 2014;25(2):225–231.
    View this article via: CrossRef PubMed Google Scholar
  140. Huugen D, et al. Inhibition of complement factor C5 protects against anti-myeloperoxidase antibody-mediated glomerulonephritis in mice. Kidney Int. 2007;71(7):646–654.
    View this article via: CrossRef PubMed Google Scholar
  141. Poppelaars F, Thurman JM. Complement-mediated kidney diseases. Mol Immunol. 2020;128:175–187.
    View this article via: CrossRef PubMed Google Scholar
  142. Stühlinger W, et al. Letter: Haemolytic-uraemic syndrome: evidence for intravascular C3 activation. Lancet. 1974;2(7883):788–789.
    View this article via: CrossRef PubMed Google Scholar
  143. Drukker A, et al. Recurrent hemolytic-uremic syndrome: a case report. Clin Nephrol. 1975;4(2):68–72.
    View this article via: PubMed CrossRef Google Scholar
  144. Kourilsky O, et al. Persistent intravascular C3 activation after bilateral nephrectomy in patients with thrombotic microangiopathy. Clin Nephrol. 1976;6(4):437–439.
    View this article via: PubMed Google Scholar
  145. Kim Y, et al. Breakdown products of C3 and factor B in hemolytic-uremic syndrome. J Lab Clin Med. 1977;89(4):845–850.
    View this article via: PubMed Google Scholar
  146. Warwicker P, et al. Genetic studies into inherited and sporadic hemolytic uremic syndrome. Kidney Int. 1998;53(4):836–844.
    View this article via: CrossRef PubMed Google Scholar
  147. Thompson RA, Winterborn MH. Hypocomplementaemia due to a genetic deficiency of beta 1H globulin. Clin Exp Immunol. 1981;46(1):110–119.
    View this article via: PubMed Google Scholar
  148. Roodhooft AM, et al. Recurrent haemolytic uraemic syndrome and acquired hypomorphic variant of the third component of complement. Pediatr Nephrol. 1990;4(6):597–599.
    View this article via: CrossRef PubMed Google Scholar
  149. Pichette V, et al. Familial hemolytic-uremic syndrome and homozygous factor H deficiency. Am J Kidney Dis. 1994;24(6):936–941.
    View this article via: CrossRef PubMed Google Scholar
  150. Caprioli J, et al. Genetics of HUS: the impact of MCP, CFH, and IF mutations on clinical presentation, response to treatment, and outcome. Blood. 2006;108(4):1267–1279.
    View this article via: CrossRef PubMed Google Scholar
  151. Servais A, et al. Primary glomerulonephritis with isolated C3 deposits: a new entity which shares common genetic risk factors with haemolytic uraemic syndrome. J Med Genet. 2007;44(3):193–199.
    View this article via: CrossRef PubMed Google Scholar
  152. Høgåsen K, et al. Hereditary porcine membranoproliferative glomerulonephritis type II is caused by factor H deficiency. J Clin Invest. 1995;95(3):1054–1061.
    View this article via: JCI CrossRef PubMed Google Scholar
  153. Pickering MC, et al. Uncontrolled C3 activation causes membranoproliferative glomerulonephritis in mice deficient in complement factor H. Nat Genet. 2002;31(4):424–428.
    View this article via: CrossRef PubMed Google Scholar
  154. Sánchez-Corral P, et al. Structural and functional characterization of factor H mutations associated with atypical hemolytic uremic syndrome. Am J Hum Genet. 2002;71(6):1285–1295.
    View this article via: CrossRef PubMed Google Scholar
  155. Rodríguez De Córdoba S, et al. The human complement factor H: functional roles, genetic variations and disease associations. Mol Immunol. 2004;41(4):355–367.
    View this article via: CrossRef PubMed Google Scholar
  156. Pickering MC, et al. Spontaneous hemolytic uremic syndrome triggered by complement factor H lacking surface recognition domains. J Exp Med. 2007;204(6):1249–1256.
    View this article via: CrossRef PubMed Google Scholar
  157. Pickering MC, et al. Prevention of C5 activation ameliorates spontaneous and experimental glomerulonephritis in factor H-deficient mice. Proc Natl Acad Sci U S A. 2006;103(25):9649–9654.
    View this article via: CrossRef PubMed Google Scholar
  158. Ruggenenti P, et al. C5 convertase blockade in membranoproliferative glomerulonephritis: a single-arm Clinical Trial. Am J Kidney Dis. 2019;74(2):224–238.
    View this article via: CrossRef PubMed Google Scholar
  159. Ruseva MM, et al. Loss of properdin exacerbates C3 glomerulopathy resulting from factor H deficiency. J Am Soc Nephrol. 2013;24(1):43–52.
    View this article via: CrossRef PubMed Google Scholar
  160. Lesher AM, et al. Combination of factor H mutation and properdin deficiency causes severe C3 glomerulonephritis. J Am Soc Nephrol. 2013;24(1):53–65.
    View this article via: CrossRef PubMed Google Scholar
  161. Zhang Y, et al. C3(H2O) prevents rescue of complementmediated C3 glomerulopathy in Cfh–/– Cfd–/– mice. JCI Insight. 2020;5(9):e135758.
    View this article via: JCI Insight CrossRef PubMed Google Scholar
  162. Nastuk WL, et al. Search for a neuromuscular blocking agent in the blood of patients with myasthenia gravis. Am J Med. 1959;26(3):394–409.
    View this article via: CrossRef PubMed Google Scholar
  163. Lennon VA, et al. Role of complement in the pathogenesis of experimental autoimmune myasthenia gravis. J Exp Med. 1978;147(4):973–983.
    View this article via: CrossRef PubMed Google Scholar
  164. Sahashi K, et al. Ultrastructural localization of the terminal and lytic ninth complement component (C9) at the motor end-plate in myasthenia gravis. J Neuropathol Exp Neurol. 1980;39(2):160–172.
    View this article via: CrossRef PubMed Google Scholar
  165. Fazekas A, et al. Myasthenia gravis: demonstration of membrane attack complex in muscle end-plates. Clin Neuropathol. 1986;5(2):78–83.
    View this article via: PubMed Google Scholar
  166. Tsujihata M, et al. Diagnostic significance of IgG, C3, and C9 at the limb muscle motor end-plate in minimal myasthenia gravis. Neurology. 1989;39(10):1359–1363.
    View this article via: CrossRef PubMed Google Scholar
  167. Christadoss P. C5 gene influences the development of murine myasthenia gravis. J Immunol. 1988;140(8):2589–2592.
    View this article via: CrossRef PubMed Google Scholar
  168. Biesecker G, Gomez CM. Inhibition of acute passive transfer experimental autoimmune myasthenia gravis with Fab antibody to complement C6. J Immunol. 1989;142(8):2654–2659.
    View this article via: CrossRef PubMed Google Scholar
  169. Albazli K, et al. Complement inhibitor therapy for myasthenia gravis. Front Immunol. 2020;11:917.
    View this article via: CrossRef PubMed Google Scholar
  170. Howard JF, et al. Safety and efficacy of eculizumab in anti-acetylcholine receptor antibody-positive refractory generalised myasthenia gravis (REGAIN): a phase 3, randomised, double-blind, placebo-controlled, multicentre study. Lancet Neurol. 2017;16(12):976–986.
    View this article via: CrossRef PubMed Google Scholar
  171. Muppidi S, et al. Long-term safety and efficacy of eculizumab in generalized myasthenia gravis. Muscle Nerve. 2019;60(1):14–24.
    View this article via: CrossRef PubMed Google Scholar
  172. Howard JF, et al. Safety and efficacy of zilucoplan in patients with generalised myasthenia gravis (RAISE): a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Neurol. 2023;22(5):395–406.
    View this article via: CrossRef PubMed Google Scholar
  173. Vu T, et al. Terminal complement inhibitor ravulizumab in generalized myasthenia gravis. NEJM Evid. 2022;1(5):EVIDoa2100066.
    View this article via: CrossRef PubMed Google Scholar
  174. Lennon PVA, et al. A serum autoantibody marker of neuromyelitis optica: distinction from multiple sclerosis. Lancet. 2004;364(9451):2106–2112.
    View this article via: CrossRef PubMed Google Scholar
  175. Pittock SJ, et al. Eculizumab in aquaporin-4-positive neuromyelitis optica spectrum disorder. N Engl J Med. 2019;381(7):614–625.
    View this article via: CrossRef PubMed Google Scholar
  176. Pittock SJ, et al. Ravulizumab in aquaporin-4-positive neuromyelitis optica spectrum disorder. Ann Neurol. 2023;93(6):1053–1068.
    View this article via: CrossRef PubMed Google Scholar
  177. Bradl M, et al. Neuromyelitis optica: pathogenicity of patient immunoglobulin in vivo. Ann Neurol. 2009;66(5):630–643.
    View this article via: CrossRef PubMed Google Scholar
  178. Bennett JL, et al. Intrathecal pathogenic anti-aquaporin-4 antibodies in early neuromyelitis optica. Ann Neurol. 2009;66(5):617–629.
    View this article via: CrossRef PubMed Google Scholar
  179. Kinoshita M, et al. Neuromyelitis optica: Passive transfer to rats by human immunoglobulin. Biochem Biophys Res Commun. 2009;386(4):623–627.
    View this article via: CrossRef PubMed Google Scholar
  180. Saadoun S, et al. Intra-cerebral injection of neuromyelitis optica immunoglobulin G and human complement produces neuromyelitis optica lesions in mice. Brain. 2010;133(pt 2):349–361.
    View this article via: CrossRef PubMed Google Scholar
  181. Zhang H, Verkman AS. Eosinophil pathogenicity mechanisms and therapeutics in neuromyelitis optica. J Clin Invest. 2013;123(5):2306–2316.
    View this article via: JCI CrossRef PubMed Google Scholar
  182. Zhang H, Verkman AS. Longitudinally extensive NMO spinal cord pathology produced by passive transfer of NMO-IgG in mice lacking complement inhibitor CD59. J Autoimmun. 2014;53:67–77.
    View this article via: CrossRef PubMed Google Scholar
  183. Asavapanumas N, et al. Experimental mouse model of optic neuritis with inflammatory demyelination produced by passive transfer of neuromyelitis optica-immunoglobulin G. J Neuroinflammation. 2014;11:16.
    View this article via: CrossRef PubMed Google Scholar
  184. Ratelade J, Verkman AS. Neuromyelitis optica: aquaporin-4 based pathogenesis mechanisms and new therapies. Int J Biochem Cell Biol. 2012;44(9):1519–1530.
    View this article via: CrossRef PubMed Google Scholar
  185. Uzawa A, et al. NMOSD and MOGAD: an evolving disease spectrum. Nat Rev Neurol. 2024;20(10):602–619.
    View this article via: CrossRef PubMed Google Scholar
  186. Tradtrantip L, et al. Potential therapeutic benefit of C1-esterase inhibitor in neuromyelitis optica evaluated in vitro and in an experimental rat model. PLoS One. 2014;9(9):e106824.
    View this article via: CrossRef PubMed Google Scholar
  187. Duan T, et al. Complement-dependent bystander injury to neurons in AQP4-IgG seropositive neuromyelitis optica. J Neuroinflammation. 2018;15(1):294.
    View this article via: CrossRef PubMed Google Scholar
  188. Phuan PW, et al. C1q-targeted monoclonal antibody prevents complement-dependent cytotoxicity and neuropathology in in vitro and mouse models of neuromyelitis optica. Acta Neuropathol. 2013;125(6):829–840.
    View this article via: CrossRef PubMed Google Scholar
  189. Smith PK, et al. Effects of C5 complement inhibitor pexelizumab on outcome in high-risk coronary artery bypass grafting: combined results from the PRIMO-CABG I and II trials. J Thorac Cardiovasc Surg. 2011;142(1):89–98.
    View this article via: CrossRef PubMed Google Scholar
  190. Vogel CW. The role of complement in myocardial infarction reperfusion injury: an underappreciated therapeutic target. Front Cell Dev Biol. 2020;8:606407.
    View this article via: CrossRef PubMed Google Scholar
  191. Diepenhorst GMP, et al. Complement-mediated ischemia-reperfusion injury: lessons learned from animal and clinical studies. Ann Surg. 2009;249(6):889–899.
    View this article via: CrossRef PubMed Google Scholar
  192. Granger CB, et al. Pexelizumab, an anti-C5 complement antibody, as adjunctive therapy to primary percutaneous coronary intervention in acute myocardial infarction: the COMplement inhibition in Myocardial infarction treated with Angioplasty (COMMA) trial. Circulation. 2003;108(10):1184–1190.
    View this article via: CrossRef PubMed Google Scholar
  193. Shernan SK, et al. Impact of pexelizumab, an anti-C5 complement antibody, on total mortality and adverse cardiovascular outcomes in cardiac surgical patients undergoing cardiopulmonary bypass. Ann Thorac Surg. 2004;77(3):942–949.
    View this article via: CrossRef PubMed Google Scholar
  194. Armstrong PW, et al. Concerning the mechanism of pexelizumab’s benefit in acute myocardial infarction. Am Heart J. 2006;151(4):787–790.
    View this article via: CrossRef PubMed Google Scholar
  195. Fitch JCK, et al. Pharmacology and biological efficacy of a recombinant, humanized, single-chain antibody C5 complement inhibitor in patients undergoing coronary artery bypass graft surgery with cardiopulmonary bypass. Circulation. 1999;100(25):2499–2506.
    View this article via: CrossRef PubMed Google Scholar
  196. Mahaffey KW, et al. Effect of pexelizumab, an anti-C5 complement antibody, as adjunctive therapy to fibrinolysis in acute myocardial infarction: the COMPlement inhibition in myocardial infarction treated with thromboLYtics (COMPLY) trial. Circulation. 2003;108(10):1176–1183.
    View this article via: CrossRef PubMed Google Scholar
  197. Armstrong PW, et al. Pexelizumab for acute ST-elevation myocardial infarction in patients undergoing primary percutaneous coronary intervention: a randomized controlled trial. JAMA. 2007;297(1):43–51.
    View this article via: CrossRef PubMed Google Scholar
  198. Verrier ED, et al. Terminal complement blockade with pexelizumab during coronary artery bypass graft surgery requiring cardiopulmonary bypass: a randomized trial. JAMA. 2004;291(19):2319–2327.
    View this article via: CrossRef PubMed Google Scholar
  199. Lazar HL, et al. Soluble human complement receptor 1 limits ischemic damage in cardiac surgery patients at high risk requiring cardiopulmonary bypass. Circulation. 2004;110(11 suppl 1):II274–II279.
    View this article via: PubMed CrossRef Google Scholar
  200. Weisman HF, et al. Soluble human complement receptor type 1: in vivo inhibitor of complement suppressing post-ischemic myocardial inflammation and necrosis. Science. 1990;249(4965):146–151.
    View this article via: CrossRef PubMed Google Scholar
  201. Eikelboom JW, O’Donnell M. Pexelizumab does not “complement” percutaneous coronary intervention in patients with ST-elevation myocardial infarction. JAMA. 2007;297(1):91–92.
    View this article via: CrossRef PubMed Google Scholar
  202. Lin GM, et al. A critical appraisal of pexelizumab treatment in patients undergoing coronary artery bypass grafting. J Thorac Cardiovasc Surg. 2011;142(2):472; author reply 473.
    View this article via: CrossRef PubMed Google Scholar
  203. Leonardi S, et al. Implementation of standardized assessment and reporting of myocardial infarction in contemporary randomized controlled trials: a systematic review. Eur Heart J. 2013;34(12):894–902.
    View this article via: CrossRef PubMed Google Scholar
  204. Testa L, et al. Pexelizumab in ischemic heart disease: a systematic review and meta-analysis on 15,196 patients. J Thorac Cardiovasc Surg. 2008;136(4):884–893.
    View this article via: CrossRef PubMed Google Scholar
  205. Lazar HL, et al. Beneficial effects of complement inhibition with soluble complement receptor 1 (TP10) during cardiac surgery: is there a gender difference? Circulation. 2007;116(11 suppl):I83–I88.
    View this article via: PubMed CrossRef Google Scholar
  206. Schwartz Longacre L, et al. New horizons in cardioprotection: Recommendations from the 2010 National Heart, Lung, and Blood Institute workshop. Circulation. 2011;124(10):1172–1179.
    View this article via: CrossRef PubMed Google Scholar
  207. Girardi G, et al. Heparin prevents antiphospholipid antibody-induced fetal loss by inhibiting complement activation. Nat Med. 2004;10(11):1222–1226.
    View this article via: CrossRef PubMed Google Scholar
  208. Steggerda JA, Heeger PS. The promise of complement therapeutics in solid organ transplantation. Transplantation. 2024;108(9):1882–1894.
    View this article via: CrossRef PubMed Google Scholar
  209. Wang H, et al. Prevention of acute vascular rejection by a functionally blocking anti-C5 monoclonal antibody combined with cyclosporine. Transplantation. 2005;79(9):1121–1127.
    View this article via: CrossRef PubMed Google Scholar
  210. Wang H, et al. Inhibition of terminal complement components in presensitized transplant recipients prevents antibody-mediated rejection leading to long-term graft survival and accommodation. J Immunol. 2007;179(7):4451–4463.
    View this article via: CrossRef PubMed Google Scholar
  211. Rother RP, et al. C5 blockade with conventional immunosuppression induces long-term graft survival in presensitized recipients. Am J Transplant. 2008;8(6):1129–1142.
    View this article via: CrossRef PubMed Google Scholar
  212. Tillou X, et al. Recombinant human C1-inhibitor prevents acute antibody-mediated rejection in alloimmunized baboons. Kidney Int. 2010;78(2):152–159.
    View this article via: CrossRef PubMed Google Scholar
  213. Eerhart MJ, et al. Complement blockade in recipients prevents delayed graft function and delays antibody-mediated rejection in a nonhuman primate model of kidney transplantation. Transplantation. 2022;106(1):60–71.
    View this article via: CrossRef PubMed Google Scholar
  214. Stegall MD, et al. Terminal complement inhibition decreases antibody-mediated rejection in sensitized renal transplant recipients. Am J Transplant. 2011;11(11):2405–2413.
    View this article via: CrossRef PubMed Google Scholar
  215. Orandi BJ, et al. Eculizumab and splenectomy as salvage therapy for severe antibody-mediated rejection after HLA-incompatible kidney transplantation. Transplantation. 2014;98(8):857–863.
    View this article via: CrossRef PubMed Google Scholar
  216. Cornell LD, et al. Positive crossmatch kidney transplant recipients treated with eculizumab: outcomes beyond 1 year. Am J Transplant. 2015;15(5):1293–1302.
    View this article via: CrossRef PubMed Google Scholar
  217. Kulkarni S, et al. Eculizumab therapy for chronic antibody-mediated injury in kidney transplant recipients: a pilot randomized controlled trial. Am J Transplant. 2017;17(3):682–691.
    View this article via: CrossRef PubMed Google Scholar
  218. Marks WH, et al. Safety and efficacy of eculizumab in the prevention of antibody-mediated rejection in living-donor kidney transplant recipients requiring desensitization therapy: A randomized trial. Am J Transplant. 2019;19(10):2876–2888.
    View this article via: CrossRef PubMed Google Scholar
  219. Schinstock CA, et al. Long-term outcomes of eculizumab-treated positive crossmatch recipients: Allograft survival, histologic findings, and natural history of the donor-specific antibodies. Am J Transplant. 2019;19(6):1671–1683.
    View this article via: CrossRef PubMed Google Scholar
  220. Vo AA, et al. A phase I/II placebo-controlled trial of C1-inhibitor for prevention of antibody-mediated rejection in HLA sensitized patients. Transplantation. 2015;99(2):299–308.
    View this article via: CrossRef PubMed Google Scholar
  221. Montgomery RA, et al. Plasma-derived C1 esterase inhibitor for acute antibody-mediated rejection following kidney transplantation: results of a randomized double-blind placebo-controlled pilot study. Am J Transplant. 2016;16(12):3468–3478.
    View this article via: CrossRef PubMed Google Scholar
  222. Viglietti D, et al. C1 inhibitor in acute antibody-mediated rejection nonresponsive to conventional therapy in kidney transplant recipients: a pilot study. Am J Transplant. 2016;16(5):1596–1603.
    View this article via: CrossRef PubMed Google Scholar
  223. Golshayan D, et al. Targeting the complement pathway in kidney transplantation. J Am Soc Nephrol. 2023;34(11):1776–1792.
    View this article via: CrossRef PubMed Google Scholar
  224. Eskandary F, et al. Anti-C1s monoclonal antibody BIVV009 in late antibody-mediated kidney allograft rejection-results from a first-in-patient phase 1 trial. Am J Transplant. 2018;18(4):916–926.
    View this article via: CrossRef PubMed Google Scholar
  225. Jordan SC, et al. A phase I/II, double-blind, placebo-controlled study assessing safety and efficacy of C1 esterase inhibitor for prevention of delayed graft function in deceased donor kidney transplant recipients. Am J Transplant. 2018;18(12):2955–2964.
    View this article via: CrossRef PubMed Google Scholar
  226. Schröppel B, et al. Peritransplant eculizumab does not prevent delayed graft function in deceased donor kidney transplant recipients: Results of two randomized controlled pilot trials. Am J Transplant. 2020;20(2):564–572.
    View this article via: CrossRef PubMed Google Scholar
  227. Huang E, et al. Three-year outcomes of a randomized, double-blind, placebo-controlled study assessing safety and efficacy of C1 esterase inhibitor for prevention of delayed graft function in deceased donor kidney transplant recipients. Clin J Am Soc Nephrol. 2020;15(1):109–116.
    View this article via: CrossRef PubMed Google Scholar
  228. Sommer W, et al. C1-esterase-inhibitor for primary graft dysfunction in lung transplantation. Transplantation. 2014;97(11):1185–1191.
    View this article via: CrossRef PubMed Google Scholar
  229. Kassimatis T, et al. A double-blind randomised controlled investigation into the efficacy of Mirococept (APT070) for preventing ischaemia reperfusion injury in the kidney allograft (EMPIRIKAL): study protocol for a randomised controlled trial. Trials. 2017;18(1):255.
    View this article via: CrossRef PubMed Google Scholar
  230. Meurisse N, et al. Effect of a combined drug approach on the severity of ischemia-reperfusion injury during liver transplant: a randomized clinical trial. JAMA Netw Open. 2023;6(2):e230819.
    View this article via: CrossRef PubMed Google Scholar
  231. Zhou W, et al. Predominant role for C5b-9 in renal ischemia/reperfusion injury. J Clin Invest. 2000;105(10):1363–1371.
    View this article via: JCI CrossRef PubMed Google Scholar
  232. De Vries B, et al. Inhibition of complement factor C5 protects against renal ischemia-reperfusion injury: inhibition of late apoptosis and inflammation. Transplantation. 2003;75(3):375–382.
    View this article via: CrossRef PubMed Google Scholar
  233. Castellano G, et al. Therapeutic targeting of classical and lectin pathways of complement protects from ischemia-reperfusion-induced renal damage. Am J Pathol. 2010;176(4):1648–1659.
    View this article via: CrossRef PubMed Google Scholar
  234. Yu ZX, et al. Targeting complement pathways during cold ischemia and reperfusion prevents delayed graft function. Am J Transplant. 2016;16(9):2589–2597.
    View this article via: CrossRef PubMed Google Scholar
  235. Holguin MH, et al. Relationship between the membrane inhibitor of reactive lysis and the erythrocyte phenotypes of paroxysmal nocturnal hemoglobinuria. J Clin Invest. 1989;84(5):1387–1394.
    View this article via: JCI CrossRef PubMed Google Scholar
  236. Chen Y, Rong F. Advances in the creation of animal models of paroxysmal nocturnal hemoglobinuria. Hematology. 2021;26(1):491–496.
    View this article via: CrossRef PubMed Google Scholar
  237. Risitano AM, et al. The complement receptor 2/factor H fusion protein TT30 protects paroxysmal nocturnal hemoglobinuria erythrocytes from complement-mediated hemolysis and C3 fragment. Blood. 2012;119(26):6307–6316.
    View this article via: CrossRef PubMed Google Scholar
  238. Rosse WF. Fixation of the first component of complement (C’la) by human antibodies. J Clin Invest. 1969;47(11):2430–2445.
    View this article via: JCI CrossRef PubMed Google Scholar
  239. Berentsen S. Complement activation and inhibition in autoimmune hemolytic anemia: focus on cold agglutinin disease. Semin Hematol. 2018;55(3):141–149.
    View this article via: CrossRef PubMed Google Scholar
  240. Howie HL, Hudson KE. Murine models of autoimmune hemolytic anemia. Curr Opin Hematol. 2018;25(6):473–481.
    View this article via: CrossRef PubMed Google Scholar
  241. Shi J, et al. TNT003, an inhibitor of the serine protease C1s, prevents complement activation induced by cold agglutinins. Blood. 2014;123(26):4015–4022.
    View this article via: CrossRef PubMed Google Scholar
  242. Röth A, et al. Sutimlimab in cold agglutinin disease. N Engl J Med. 2021;384(14):1323–1334.
    View this article via: CrossRef PubMed Google Scholar
  243. Jaffe CJ, et al. The role of complement in the clearance of cold agglutinin-sensitized erythrocytes in man. J Clin Invest. 1976;58(4):942–949.
    View this article via: JCI CrossRef PubMed Google Scholar
  244. Klein RJ, et al. Complement factor H polymorphism in age-related macular degeneration. Science. 2005;308(5720):385–389.
    View this article via: CrossRef PubMed Google Scholar
  245. Edwards AO, et al. Complement factor H polymorphism and age-related macular degeneration. Science. 2005;308(5720):421–424.
    View this article via: CrossRef PubMed Google Scholar
  246. Haines JL, et al. Complement factor H variant increases the risk of age-related macular degeneration. Science. 2005;308(5720):419–421.
    View this article via: CrossRef PubMed Google Scholar
  247. Gold B, et al. Variation in factor B (BF) and complement component 2 (C2) genes is associated with age-related macular degeneration. Nat Genet. 2006;38(4):458–462.
    View this article via: CrossRef PubMed Google Scholar
  248. Yates JRW, et al. Complement C3 variant and the risk of age-related macular degeneration. N Engl J Med. 2007;357(6):553–561.
    View this article via: CrossRef PubMed Google Scholar
  249. Liao DS, et al. Complement C3 inhibitor pegcetacoplan for geographic atrophy secondary to age-related macular degeneration: a randomized phase 2 trial. Ophthalmology. 2020;127(2):186–195.
    View this article via: CrossRef PubMed Google Scholar
  250. Nittala MG, et al. Association of pegcetacoplan with progression of incomplete retinal pigment epithelium and outer retinal atrophy in age-related macular degeneration: a post hoc analysis of the FILLY Randomized Clinical Trial. JAMA Ophthalmol. 2022;140(3):243–249.
    View this article via: CrossRef PubMed Google Scholar
  251. Jaffe GJ, et al. C5 inhibitor avacincaptad pegol for geographic atrophy due to age-related macular degeneration: A Randomized Pivotal Phase 2/3 Trial. Ophthalmology. 2021;128(4):576–586.
    View this article via: CrossRef PubMed Google Scholar
  252. Coffey PJ, et al. Complement factor H deficiency in aged mice causes retinal abnormalities and visual dysfunction. Proc Natl Acad Sci U S A. 2007;104(42):16651–16656.
    View this article via: CrossRef PubMed Google Scholar
  253. Ufret-Vincenty RL, et al. Transgenic mice expressing variants of complement factor H develop AMD-like retinal findings. Invest Ophthalmol Vis Sci. 2010;51(11):5878–5887.
    View this article via: CrossRef PubMed Google Scholar
  254. Cashman SM, et al. Expression of complement component 3 (C3) from an adenovirus leads to pathology in the murine retina. Invest Ophthalmol Vis Sci. 2011;52(6):3436–3445.
    View this article via: CrossRef PubMed Google Scholar
  255. Toomey CB, et al. Regulation of age-related macular degeneration-like pathology by complement factor H. Proc Natl Acad Sci U S A. 2015;112(23):E3040–E3049.
    View this article via: CrossRef PubMed Google Scholar
  256. Lyzogubov VV, et al. The complement regulatory protein CD46 deficient mouse spontaneously develops dry-type age-related macular degeneration-like phenotype. Am J Pathol. 2016;186(8):2088–2104.
    View this article via: CrossRef PubMed Google Scholar
  257. Landowski M, et al. Human complement factor H Y402H polymorphism causes an age-related macular degeneration phenotype and lipoprotein dysregulation in mice. Proc Natl Acad Sci U S A. 2019;116(9):3703–3711.
    View this article via: CrossRef PubMed Google Scholar
  258. Feng L, et al. Complement factor H deficiency combined with smoking promotes retinal degeneration in a novel mouse model. Exp Biol Med (Maywood). 2022;247(2):77–86.
    View this article via: CrossRef PubMed Google Scholar
  259. Perkovic V, et al. Alternative complement pathway inhibition with Iptacopan in IgA nephropathy. N Engl J Med. 2024;392(6):531–543.
    View this article via: CrossRef PubMed Google Scholar
  260. Zhang Y, et al. Targeting C3a/C5a receptors inhibits human mesangial cell proliferation and alleviates immunoglobulin A nephropathy in mice. Clin Exp Immunol. 2017;189(1):60–70.
    View this article via: CrossRef PubMed Google Scholar
  261. Poppelaars F, et al. The contribution of complement to the pathogenesis of IgA nephropathy: are complement-targeted therapies moving from rare disorders to more common diseases? J Clin Med. 2021;10(20):4715.
    View this article via: CrossRef PubMed Google Scholar
  262. Ozen A, et al. CD55 deficiency, early-onset protein-losing enteropathy, and thrombosis. N Engl J Med. 2017;377(1):52–61.
    View this article via: CrossRef PubMed Google Scholar
  263. Kurolap A, et al. Loss of CD55 in eculizumab-responsive protein-losing enteropathy. N Engl J Med. 2017;377(1):87–89.
    View this article via: CrossRef PubMed Google Scholar
  264. Ozen A, et al. Evaluating the efficacy and safety of pozelimab in patients with CD55 deficiency with hyperactivation of complement, angiopathic thrombosis, and protein-losing enteropathy disease: an open-label phase 2 and 3 study. Lancet. 2024;403(10427):645–656.
    View this article via: CrossRef PubMed Google Scholar
  265. Miwa T, et al. Deletion of decay-accelerating factor (CD55) exacerbates autoimmune disease development in MRL/lpr mice. Am J Pathol. 2002;161(3):1077–1086.
    View this article via: CrossRef PubMed Google Scholar
  266. Miwa T, et al. Decay-accelerating factor ameliorates systemic autoimmune disease in MRL/lpr mice via both complement-dependent and -independent mechanisms. Am J Pathol. 2007;170(4):1258–1266.
    View this article via: CrossRef PubMed Google Scholar
  267. Lin F, et al. Decay-accelerating factor deficiency increases susceptibility to dextran sulfate sodium-induced colitis: role for complement in inflammatory bowel disease. J Immunol. 2004;172(6):3836–3841.
    View this article via: CrossRef PubMed Google Scholar
  268. Liu J, et al. The complement inhibitory protein DAF (CD55) suppresses T cell immunity in vivo. J Exp Med. 2005;201(4):567–577.
    View this article via: CrossRef PubMed Google Scholar
  269. Minikel EV, et al. Refining the impact of genetic evidence on clinical success. Nature. 2024;629(8012):624–629.
    View this article via: CrossRef PubMed Google Scholar
  270. Ineichen BV, et al. Analysis of animal-to-human translation shows that only 5% of animal-tested therapeutic interventions obtain regulatory approval for human applications. PLoS Biol. 2024;22(6):e3002667.
    View this article via: CrossRef PubMed Google Scholar
  271. van Damme BJC, et al. Experimental glomerulonephritis in the rat induced by antibodies directed against tubular antigens. V. Fixed glomerular antigens in the pathogenesis of heterologous immune complex glomerulonephritis. Lab Invest. 1978;38(4):502–510.
    View this article via: PubMed Google Scholar
  272. Barratt J, et al. Phase 2 trial of cemdisiran in adult patients with IgA nephropathy: a randomized controlled trial. Clin J Am Soc Nephrol. 2024;19(4):452–462.
    View this article via: CrossRef PubMed Google Scholar
  273. Appel G, et al. Eculizamab (C5 complement inhibitor) in the treatment of idiopathic membranous nephropathy. J Am Soc Nephrol. 2002;13:668A.
  274. Ma H, et al. The role of complement in membranous nephropathy. Semin Nephrol. 2013;33(6):531–542.
    View this article via: CrossRef PubMed Google Scholar
  275. Cunningham PN, Quigg RJ. Contrasting roles of complement activation and its regulation in membranous nephropathy. J Am Soc Nephrol. 2005;16(5):1214–1222.
    View this article via: CrossRef PubMed Google Scholar
  276. Ter Avest M, et al. Proteinuria and exposure to eculizumab in atypical hemolytic uremic syndrome. Clin J Am Soc Nephrol. 2023;18(6):759–766.
    View this article via: CrossRef PubMed Google Scholar
  277. Vergunst CE, et al. Blocking the receptor for C5a in patients with rheumatoid arthritis does not reduce synovial inflammation. Rheumatology (Oxford). 2007;46(12):1773–1778.
    View this article via: CrossRef PubMed Google Scholar
  278. Mojcik CF, et al. Results of a phase 2b study of the humanized anti-C5 antibody eculizumab in patients with rheumatoid arthritis. Ann Rheum Dis. 2004;63(suppl 1):301.
  279. Renner B, et al. Noninvasive detection of iC3b/C3d deposits in the kidney using a novel bioluminescent imaging probe. J Am Soc Nephrol. 2023;34(7):1151–1154.
    View this article via: CrossRef PubMed Google Scholar
  280. Markiewski MM, et al. Modulation of the antitumor immune response by complement. Nat Immunol. 2008;9(11):1225–1235.
    View this article via: CrossRef PubMed Google Scholar
  281. Laskowski J, et al. Complement factor H-deficient mice develop spontaneous hepatic tumors. J Clin Invest. 2020;130(8):4039–4054.
    View this article via: JCI PubMed CrossRef Google Scholar
  282. Petry F, et al. The mouse C1q genes are clustered on chromosome 4 and show conservation of gene organization. Immunogenetics. 1996;43(6):370–376.
    View this article via: CrossRef PubMed Google Scholar
  283. Arkwright PD, et al. Successful cure of C1q deficiency in human subjects treated with hematopoietic stem cell transplantation. J Allergy Clin Immunol. 2014;133(1):265–267.
    View this article via: CrossRef PubMed Google Scholar
  284. Petry F, et al. Reconstitution of the complement function in C1q-deficient (C1qa-/-) mice with wild-type bone marrow cells. J Immunol. 2001;167(7):4033–4037.
    View this article via: CrossRef PubMed Google Scholar
  285. Botto M, et al. Homozygous C1q deficiency causes glomerulonephritis associated with multiple apoptotic bodies. Nat Genet. 1998;19(1):56–59.
    View this article via: CrossRef PubMed Google Scholar
  286. Webster SD, et al. The mouse C1q A-chain sequence alters beta-amyloid-induced complement activation. Neurobiol Aging. 1999;20(3):297–304.
    View this article via: CrossRef PubMed Google Scholar
  287. Lawson PR, Reid KBM. A novel PCR-based technique using expressed sequence tags and gene homology for murine genetic mapping: localization of the complement genes. Int Immunol. 2000;12(3):231–240.
    View this article via: CrossRef PubMed Google Scholar
  288. Fujita T, et al. The lectin-complement pathway--its role in innate immunity and evolution. Immunol Rev. 2004;198:185–202.
    View this article via: CrossRef PubMed Google Scholar
  289. Liu H, et al. Characterization and quantification of mouse mannan-binding lectins (MBL-A and MBL-C) and study of acute phase responses. Scand J Immunol. 2001;53(5):489–497.
    View this article via: CrossRef PubMed Google Scholar
  290. Hansen S, et al. Purification and characterization of two mannan-binding lectins from mouse serum. J Immunol. 2000;164(5):2610–2618.
    View this article via: CrossRef PubMed Google Scholar
  291. Garred P, et al. MBL2, FCN1, FCN2 and FCN3-The genes behind the initiation of the lectin pathway of complement. Mol Immunol. 2009;46(14):2737–2744.
    View this article via: CrossRef PubMed Google Scholar
  292. Endo Y, et al. Mouse ficolin B has an ability to form complexes with mannose-binding lectin-associated serine proteases and activate complement through the lectin pathway. J Biomed Biotechnol. 2012;2012:105891.
    View this article via: CrossRef PubMed Google Scholar
  293. Endo Y, et al. Carbohydrate-binding specificities of mouse ficolin A, a splicing variant of ficolin A and ficolin B and their complex formation with MASP-2 and sMAP. Immunogenetics. 2005;57(11):837–844.
    View this article via: CrossRef PubMed Google Scholar
  294. Rooryck C, et al. Mutations in lectin complement pathway genes COLEC11 and MASP1 cause 3MC syndrome. Nat Genet. 2011;43(3):197–203.
    View this article via: CrossRef PubMed Google Scholar
  295. Farrar CA, et al. Collectin-11 detects stress-induced L-fucose pattern to trigger renal epithelial injury. J Clin Invest. 2016;126(5):1911–1925.
    View this article via: JCI CrossRef PubMed Google Scholar
  296. Henriksen ML, et al. Heteromeric complexes of native collectin kidney 1 and collectin liver 1 are found in the circulation with MASPs and activate the complement system. J Immunol. 2013;191(12):6117–6127.
    View this article via: CrossRef PubMed Google Scholar
  297. Motomura W, et al. Immunolocalization of a novel collectin CL-K1 in murine tissues. J Histochem Cytochem. 2008;56(3):243–252.
    View this article via: CrossRef PubMed Google Scholar
  298. Héja D, et al. Revised mechanism of complement lectin-pathway activation revealing the role of serine protease MASP-1 as the exclusive activator of MASP-2. Proc Natl Acad Sci U S A. 2012;109(26):10498–10503.
    View this article via: CrossRef PubMed Google Scholar
  299. Skjoedt MO, et al. A novel mannose-binding lectin/ficolin-associated protein is highly expressed in heart and skeletal muscle tissues and inhibits complement activation. J Biol Chem. 2010;285(11):8234–8243.
    View this article via: CrossRef PubMed Google Scholar
  300. Skjoedt MO, et al. Crystal structure and functional characterization of the complement regulator mannose-binding lectin (MBL)/ficolin-associated protein-1 (MAP-1). J Biol Chem. 2012;287(39):32913–32921.
    View this article via: CrossRef PubMed Google Scholar
  301. Degn SE, et al. Co-complexes of MASP-1 and MASP-2 associated with the soluble pattern-recognition molecules drive lectin pathway activation in a manner inhibitable by MAp44. J Immunol. 2013;191(3):1334–1345.
    View this article via: CrossRef PubMed Google Scholar
  302. Sirmaci A, et al. MASP1 mutations in patients with facial, umbilical, coccygeal, and auditory findings of Carnevale, Malpuech, OSA, and Michels syndromes. Am J Hum Genet. 2010;87(5):679–686.
    View this article via: CrossRef PubMed Google Scholar
  303. Diebolder CA, et al. Complement is activated by IgG hexamers assembled at the cell surface. Science. 2014;343(6176):1260–1263.
    View this article via: CrossRef PubMed Google Scholar
  304. Lubbers R, et al. Carbamylation reduces the capacity of IgG for hexamerization and complement activation. Clin Exp Immunol. 2020;200(1):1–11.
    View this article via: CrossRef PubMed Google Scholar
  305. Quast I, et al. Sialylation of IgG Fc domain impairs complement-dependent cytotoxicity. J Clin Invest. 2015;125(11):4160–4170.
    View this article via: JCI CrossRef PubMed Google Scholar
  306. Seino J, et al. Activation of human complement by mouse and mouse/human chimeric monoclonal antibodies. Clin Exp Immunol. 1993;94(2):291–296.
    View this article via: CrossRef PubMed Google Scholar
  307. Koolwijk P, et al. Binding of the human complement subcomponent C1q to hybrid mouse monoclonal antibodies. Mol Immunol. 1991;28(6):567–576.
    View this article via: CrossRef PubMed Google Scholar
  308. Presanis JS, et al. Differential substrate and inhibitor profiles for human MASP-1 and MASP-2. Mol Immunol. 2004;40(13):921–929.
    View this article via: CrossRef PubMed Google Scholar
  309. Davis AE, et al. C1 inhibitor, a multi-functional serine protease inhibitor. Thromb Haemost. 2010;104(5):886–893.
    View this article via: PubMed CrossRef Google Scholar
  310. Bupp S, et al. A novel murine in vivo model for acute hereditary angioedema attacks. Sci Rep. 2021;11(1):15924.
    View this article via: CrossRef PubMed Google Scholar
  311. Fukuoka Y, et al. Purification of the fourth, second and fifth components of mouse complement. Immunology. 1984;51(3):493–501.
    View this article via: PubMed Google Scholar
  312. Nielsen HE, et al. Rate-limiting components and reaction steps in complement-mediated haemolysis. APMIS. 1992;100(12):1053–1060.
    View this article via: CrossRef PubMed Google Scholar
  313. Ishikawa N, et al. Murine complement C2 and factor B genomic and cDNA cloning reveals different mechanisms for multiple transcripts of C2 and B. J Biol Chem. 1990;265(31):19040–19046.
    View this article via: CrossRef PubMed Google Scholar
  314. Tange CE, et al. Quantification of human complement C2 protein using an automated turbidimetric immunoassay. Clin Chem Lab Med. 2018;56(9):1498–1506.
    View this article via: CrossRef PubMed Google Scholar
  315. Paul E, et al. Anti-DNA autoreactivity in C4-deficient mice. Eur J Immunol. 2002;32(9):2672–2679.
    View this article via: CrossRef PubMed Google Scholar
  316. Härdig Y, et al. The amino-terminal module of the C4b-binding protein alpha-chain is crucial for C4b binding and factor I-cofactor function. Biochem J. 1997;323 (pt 2):469–475.
    View this article via: CrossRef PubMed Google Scholar
  317. Kaidoh T, et al. Murine C4-binding protein: a rapid purification method by affinity chromatography. J Immunol. 1981;126(2):463–467.
    View this article via: CrossRef PubMed Google Scholar
  318. Kristensen T, et al. cDNA structure of murine C4b-binding protein, a regulatory component of the serum complement system. Biochemistry. 1987;26(15):4668–4674.
    View this article via: CrossRef PubMed Google Scholar
  319. Ermert D, Blom AM. C4b-binding protein: the good, the bad and the deadly. Novel functions of an old friend. Immunol Lett. 2016;169:82–92.
    View this article via: CrossRef PubMed Google Scholar
  320. Elvington M, et al. Evolution of the complement system: from defense of the single cell to guardian of the intravascular space. Immunol Rev. 2016;274(1):9–15.
    View this article via: CrossRef PubMed Google Scholar
  321. Thurman JM, et al. Lack of a functional alternative complement pathway ameliorates ischemic acute renal failure in mice. J Immunol. 2003;170(3):1517–1523.
    View this article via: CrossRef PubMed Google Scholar
  322. Slade C, et al. Deficiency in complement factor B. N Engl J Med. 2013;369(17):1667–1669.
    View this article via: CrossRef PubMed Google Scholar
  323. Lesavre PH, Müller-Eberhard HJ. Mechanism of action of factor D of the alternative complement pathway. J Exp Med. 1978;148(6):1498–1509.
    View this article via: CrossRef PubMed Google Scholar
  324. Volanakis JE, et al. Renal filtration and catabolism of complement protein D. N Engl J Med. 1985;312(7):395–399.
    View this article via: CrossRef PubMed Google Scholar
  325. Hiemstra PS, et al. Complete and partial deficiencies of complement factor D in a Dutch family. J Clin Invest. 1989;84(6):1957–1961.
    View this article via: JCI CrossRef PubMed Google Scholar
  326. Xu Y, et al. Complement activation in factor D-deficient mice. Proc Natl Acad Sci U S A. 2001;98(25):14577–14582.
    View this article via: CrossRef PubMed Google Scholar
  327. White RT, et al. Human adipsin is identical to complement factor D and is expressed at high levels in adipose tissue. J Biol Chem. 1992;267(13):9210–9213.
    View this article via: CrossRef PubMed Google Scholar
  328. Wu X, et al. Contribution of adipose-derived factor D/adipsin to complement alternative pathway activation: lessons from lipodystrophy. J Immunol. 2018;200(8):2786–2797.
    View this article via: CrossRef PubMed Google Scholar
  329. Lubbers R, et al. Production of complement components by cells of the immune system. Clin Exp Immunol. 2017;188(2):183–194.
    View this article via: CrossRef PubMed Google Scholar
  330. Maves KK, et al. Cloning and characterization of the cDNA encoding guinea-pig properdin: a comparison of properdin from three species. Immunology. 1995;86(3):475–479.
    View this article via: PubMed Google Scholar
  331. Pangburn MK. Analysis of the natural polymeric forms of human properdin and their functions in complement activation. J Immunol. 1989;142(1):202–207.
    View this article via: CrossRef PubMed Google Scholar
  332. Pouw RB, et al. Of mice and men: The factor H protein family and complement regulation. Mol Immunol. 2015;67(1):12–20.
    View this article via: CrossRef PubMed Google Scholar
  333. Alexander JJ, et al. Abnormal immune complex processing and spontaneous glomerulonephritis in complement factor H-deficient mice with human complement receptor 1 on erythrocytes. J Immunol. 2010;185(6):3759–3767.
    View this article via: CrossRef PubMed Google Scholar
  334. Schwaeble W, et al. Human complement factor H: expression of an additional truncated gene product of 43 kDa in human liver. Eur J Immunol. 1987;17(10):1485–1489.
    View this article via: CrossRef PubMed Google Scholar
  335. Hellwage J, et al. Two factor H-related proteins from the mouse: expression analysis and functional characterization. Immunogenetics. 2006;58(11):883–893.
    View this article via: CrossRef PubMed Google Scholar
  336. Vik DP, et al. Factor H. Curr Top Microbiol Immunol. 1990;153:147–162.
    View this article via: PubMed CrossRef Google Scholar
  337. Nilsson SC, et al. Complement factor I in health and disease. Mol Immunol. 2011;48(14):1611–1620.
    View this article via: CrossRef PubMed Google Scholar
  338. Lachmann PJ. The story of complement factor I. Immunobiology. 2019;224(4):511–517.
    View this article via: CrossRef PubMed Google Scholar
  339. Rose KL, et al. Factor I is required for the development of membranoproliferative glomerulonephritis in factor H-deficient mice. J Clin Invest. 2008;118(2):608–618.
    View this article via: JCI PubMed CrossRef Google Scholar
  340. Yun YS, et al. Cloning and characterization of the non-catalytic heavy chain of mouse complement factor I gene: structure comparison with the human homologue. Biochem Mol Biol Int. 1999;47(3):493–500.
    View this article via: PubMed CrossRef Google Scholar
  341. Michiyo O, et al. Functional properties of the allotypes of mouse complement regulatory protein, factor H: difference of compatibility of each allotype with human factor I. Mol Immunol. 1993;30(9):841–848.
    View this article via: CrossRef PubMed Google Scholar
  342. Riley-Vargas RC, et al. Targeted and restricted complement activation on acrosome-reacted spermatozoa. J Clin Invest. 2005;115(5):1241–1249.
    View this article via: JCI CrossRef PubMed Google Scholar
  343. Yamamoto H, et al. CD46: the ‘multitasker’ of complement proteins. Int J Biochem Cell Biol. 2013;45(12):2808–2820.
    View this article via: CrossRef PubMed Google Scholar
  344. Song WC, et al. Mouse decay-accelerating factor: selective and tissue-specific induction by estrogen of the gene encoding the glycosylphosphatidylinositol-anchored form. J Immunol. 1996;157(9):4166–4172.
    View this article via: CrossRef PubMed Google Scholar
  345. Holers VM. Complement receptors and the shaping of the natural antibody repertoire. Springer Semin Immunopathol. 2005;26(4):405–423.
    View this article via: CrossRef PubMed Google Scholar
  346. Fingeroth JD. Comparative structure and evolution of murine CR2. The homolog of the human C3d/EBV receptor (CD21). J Immunol. 1990;144(9):3458–3467.
    View this article via: CrossRef PubMed Google Scholar
  347. Fingeroth JD, et al. Identification of murine complement receptor type 2. Proc Natl Acad Sci U S A. 1989;86(1):242–246.
    View this article via: CrossRef PubMed Google Scholar
  348. Kinoshita T, et al. Regulatory proteins for the activated third and fourth components of complement (C3b and C4b) in mice. II. Identification and properties of complement receptor type 1 (CR1). J Immunol. 1985;134(4):2564–2570.
    View this article via: CrossRef PubMed Google Scholar
  349. Qian Y-M, et al. Identification and functional characterization of a new gene encoding the mouse terminal complement inhibitor CD59. J Immunol. 2000;165(5):2528–2534.
    View this article via: CrossRef PubMed Google Scholar
  350. Longhi MP, et al. Holding T cells in check--a new role for complement regulators? Trends Immunol. 2006;27(2):102–108.
    View this article via: CrossRef PubMed Google Scholar
  351. Qin X, et al. Generation and phenotyping of mCd59a and mCd59b double-knockout mice. Am J Hematol. 2009;84(2):65–70.
    View this article via: CrossRef PubMed Google Scholar
  352. Holt DS, et al. Targeted deletion of the CD59 gene causes spontaneous intravascular hemolysis and hemoglobinuria. Blood. 2001;98(2):442–449.
    View this article via: CrossRef PubMed Google Scholar
  353. Qin X, et al. Deficiency of the mouse complement regulatory protein mCd59b results in spontaneous hemolytic anemia with platelet activation and progressive male infertility. Immunity. 2003;18(2):217–227.
    View this article via: CrossRef PubMed Google Scholar
  354. Yu JX, et al. Molecular cloning of the C6A form cDNA of the mouse sixth complement component: functional integrity despite the absence of factor I modules. Immunogenetics. 2000;51(10):779–787.
    View this article via: CrossRef PubMed Google Scholar
  355. Würzner R, et al. Complement component C7. Assessment of in vivo synthesis after liver transplantation reveals that hepatocytes do not synthesize the majority of human C7. J Immunol. 1994;152(9):4624–4629.
    View this article via: CrossRef PubMed Google Scholar
  356. Hitsumoto Y, et al. Isolation of mouse complement component C7. J Immunol Methods. 1994;176(2):163–167.
    View this article via: CrossRef PubMed Google Scholar
  357. Hodeib S, et al. Human genetics of meningococcal infections. Hum Genet. 2020;139(6–7):961–980.
    View this article via: CrossRef PubMed Google Scholar
  358. Woehrl B, et al. Complement component 5 contributes to poor disease outcome in humans and mice with pneumococcal meningitis. J Clin Invest. 2011;121(10):3943–3953.
    View this article via: JCI CrossRef PubMed Google Scholar
  359. Yi K, et al. Development and evaluation of an improved mouse model of meningococcal colonization. Infect Immun. 2003;71(4):1849–1855.
    View this article via: CrossRef PubMed Google Scholar
  360. Tornetta MA, et al. The mouse anaphylatoxin C3a receptor: molecular cloning, genomic organization, and functional expression. J Immunol. 1997;158(11):5277–5282.
    View this article via: CrossRef PubMed Google Scholar
  361. Gerard C, Gerard NP. C5A anaphylatoxin and its seven transmembrane-segment receptor. Annu Rev Immunol. 1994;12:775–808.
    View this article via: CrossRef PubMed Google Scholar
  362. Hsu MH, et al. Cloning and functional characterization of the mouse C3a anaphylatoxin receptor gene. Immunogenetics. 1997;47(1):64–72.
    View this article via: CrossRef PubMed Google Scholar
  363. Cain SA, Monk PN. The orphan receptor C5L2 has high affinity binding sites for complement fragments C5a and C5a des-Arg(74). J Biol Chem. 2002;277(9):7165–7169.
    View this article via: CrossRef PubMed Google Scholar
  364. Karsten CM, et al. Monitoring C5aR2 expression using a floxed tdTomato-C5aR2 knock-in mouse. J Immunol. 2017;199(9):3234–3248.
    View this article via: CrossRef PubMed Google Scholar
  365. Quell KM, et al. Monitoring C3aR expression using a floxed tdTomato-C3aR reporter knock-in mouse. J Immunol. 2017;199(2):688–706.
    View this article via: CrossRef PubMed Google Scholar
  366. Karsten CM, et al. Monitoring and cell-specific deletion of C5aR1 using a novel floxed GFP-C5aR1 reporter knock-in mouse. J Immunol. 2015;194(4):1841–1855.
    View this article via: CrossRef PubMed Google Scholar
  367. Gavrilyuk V, et al. Identification of complement 5a-like receptor (C5L2) from astrocytes: characterization of anti-inflammatory properties. J Neurochem. 2005;92(5):1140–1149.
    View this article via: CrossRef PubMed Google Scholar
  368. Zarantonello A, et al. C3-dependent effector functions of complement. Immunol Rev. 2023;313:120–138.
    View this article via: CrossRef PubMed Google Scholar
Version history
  • Version 1 (June 16, 2025): Electronic publication

Article tools

  • View PDF
  • Download citation information
  • Send a comment
  • Terms of use
  • Standard abbreviations
  • Need help? Email the journal

Review Series

Complement Biology and Therapeutics

  • Role of local complement activation in kidney fibrosis and repair
    Didier Portilla et al.
  • Friend or foe: assessing the value of animal models for facilitating clinical breakthroughs in complement research
    Felix Poppelaars et al.
  • The secret life of complement: challenges and opportunities in exploring functions of the complosome in disease
    Tilo Freiwald et al.
  • The complement system and kidney cancer: pathogenesis to clinical applications
    Ravikumar Aalinkeel et al.
  • Complement’s involvement in allergic Th2 immunity: a cross-barrier perspective
    Sarah A. Thomas et al.
  • Chronic kidney disease enhances alternative pathway activity: a new paradigm
    Diana I. Jalal et al.
  • The multiverse of CD46 and oncologic interactions
    M. Kathryn Liszewski et al.

Metrics

  • Article usage
  • Citations to this article

Go to

  • Top
  • Abstract
  • Introduction
  • Animal testing in biomedical research
  • Using animals to understand the human complement system
  • Evaluating complement-targeted therapies: animal models versus clinical trials
  • Discussion and remarks
  • Supplemental material
  • Acknowledgments
  • Footnotes
  • References
  • Version history
Advertisement
Advertisement

Copyright © 2025 American Society for Clinical Investigation
ISSN: 0021-9738 (print), 1558-8238 (online)

Sign up for email alerts