4,399 results on '"COMPLEMENT factor H"'
Search Results
2. Mechanism of Action Underlying Ketamine's Antidepressant Effects: The AMPA Throughput Theory in Patients With Treatment-Resistant Major Depression
- Published
- 2024
3. From the Heart: Comparing the Effects of Spiritual and Secular Meditation on Psychophysiology, Cognition, Mental Health, and Social Functioning in Healthy Adults
- Author
-
Radboud University, Donders Institute for Brain, Cognition and Behaviour, Kuriakose Elias College Mannanam, Little Flower Institute of Social Sciences and Health (LISSAH) College, and Miguel Farias, Associate Professor
- Published
- 2024
4. Vaccination With Autologous Dendritic Cells Loaded With Autologous Tumor Lysate or Homogenate Combined With Immunomodulating Radiotherapy and/or Preleukapheresis IFN-alfa in Patients With Metastatic Melanoma: a Randomized 'Proof-of-principle' Phase II Study (ABSIDE)
- Published
- 2024
5. Endometriosis and Complement System
- Author
-
Merve Didem Eşkin Tanrıverdi, Medical Doctor
- Published
- 2024
6. Low Levels of Complement Factor H in the First Trimester of Pregnancy Are Associated with Spontaneous Preterm Birth.
- Author
-
Becerra-Mojica, Carlos Hernán, Mora-Guevara, Eliana, Parra-Saavedra, Miguel Antonio, Martínez-Vega, Ruth Aralí, Díaz-Martínez, Luis Alfonso, and Rincón-Orozco, Bladimiro
- Subjects
- *
COMPLEMENT factor H , *FIRST trimester of pregnancy , *HEALTH facilities , *ENZYME-linked immunosorbent assay , *COMPLEMENT (Immunology) , *PREGNANCY - Abstract
Preterm birth (PTB) remains a significant public health concern, and prediction is an important objective, particularly in the early stages of pregnancy. Many studies have relied on cervical characteristics in the mid-trimester, with limited results. It is therefore crucial to identify novel biomarkers to enhance the ability to identify women at risk. The complement pathway is implicated in the process of placentation, and recent proteomics studies have highlighted the potential roles of some complement proteins in the pathophysiology of PTB. To determine the association between the occurrence of spontaneous preterm birth (sPTB) and the concentration of complement C3, factor B, and factor H in the blood of pregnant women during the first trimester. This prospective cohort study included women with singleton pregnancies, both with and without a history of sPTB, from two health institutions in Bucaramanga, Colombia. The outcome was sPTB before 37 weeks. A blood sample was obtained between 11 + 0 to 13 + 6 weeks. ELISA immunoassay was performed to quantify the levels of C3, factor B, and factor H. A total of 355 patients were analyzed, with a rate of sPTB of 7.6% (27/355). The median plasma concentration for C3, factor B, and factor H were 488.3 μg/mL, 352.6 μg/mL, and 413.2 μg/mL, respectively. The median concentration of factor H was found to be significantly lower in patients who delivered preterm compared to patients who delivered at term (382 μg/mL vs. 415 μg/mL; p = 0.034). This study identified a significant association between low first-trimester levels of factor H and sPTB before 37 weeks. These results provide relevant information about a new possible early biomarker for sPTB. However, the results must be confirmed in different settings, and the predictive value must be examined [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Role of factor H-related protein 3 in Pseudomonas aeruginosa bloodstream infections.
- Author
-
González-Alsina, Alex, Martín-Merinero, Héctor, Mateu-Borrás, Margalida, Verd, María, Doménech-Sánchez, Antonio, Goldberg, Joanna B., Rodríguez de Córdoba, Santiago, and Albertí, Sebastián
- Subjects
PSEUDOMONAS aeruginosa infections ,COMPLEMENT factor H ,BLOOD proteins ,BACTERIAL cell surfaces ,COMPLEMENT activation - Abstract
Pseudomonas aeruginosa is a leading cause of nosocomial bloodstream infections. The outcome of these infections depends on the virulence of the microorganism as well as host-related conditions and factors. The complement system plays a crucial role in defense against bloodstream infections. P. aeruginosa counteracts complement attack by recruiting Factor H (FH) that inhibits complement amplification on the bacterial surface. Factor H-related proteins (FHRs) are a group of plasma proteins evolutionarily related to FH that have been postulated to interfere this bacterial evasion mechanism. In this study, we demonstrate that FHR-3 competes with purified FH for binding to P. aeruginosa and identify EF-Tu as a common bacterial target for both complement regulator factors. Importantly, elevated levels of FHR-3 in human serum promote complement activation, leading to increased opsonization and killing of P. aeruginosa. Conversely, physiological concentrations of FHR-3 have no significant effect. Our findings suggest that FHR-3 may serve as a protective host factor against P. aeruginosa infections. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. The SCR-17 and SCR-18 glycans in human complement factor H enhance its regulatory function.
- Author
-
Xin Gao, Iqbal, Hina, Ding-Quan Yu, Gor, Jayesh, Coker, Alun R., and Perkins, Stephen J.
- Subjects
- *
COMPLEMENT factor H , *PICHIA pastoris , *CELL membranes , *ULTRACENTRIFUGATION , *MASS spectrometry , *GLYCANS - Abstract
Human complement factor H (CFH) plays a central role in regulating activated C3b to protect host cells. CFH contain 20 short complement regulator (SCR) domains and eight N-glycosylation sites. The N-terminal SCR domains mediate C3b degradation while the C-terminal CFH domains bind to host cell surfaces to protect these. Our earlier study of Pichia-generated CFH fragments indicated a self-association site at SCR-17/18 that comprises a dimerization site for human factor H. Two N-linked glycans are located on SCR-17 and SCR-18. Here, when we expressed SCR-17/18 without glycans in an Escherichia coli system, analytical ultracentrifugation showed that no dimers were now formed. To investigate this novel finding, full-length CFH and its C-terminal fragments were purified from human plasma and Pichia pastoris respectively, and their glycans were enzymatically removed using PNGase F. Using size-exclusion chromatography, mass spectrometry, and analytical ultracentrifugation, SCR-17/18 from Pichia showed notably less dimer formation without its glycans, confirming that the glycans are necessary for the formation of SCR-17/18 dimers. By surface plasmon resonance, affinity analyses interaction showed decreased binding of deglycosylated full-length CFH to immobilized C3b, showing that CFH glycosylation enhances the key CFH regulation of C3b. We conclude that our study revealed a significant new aspect of CFH regulation based on its glycosylation and its resulting dimerization. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Enhancement of complement-dependent cytotoxicity by linking factor-H derived short consensus repeats 19-20 to CD20 antibodies.
- Author
-
Prantl, Lena, Heider, Philipp, Bergmeister, Lisa, Calana, Katharina, Bohn, Jan-Paul, Wolf, Dominik, Banki, Zoltan, Bosch, Andreas, Plach, Maximilian, Huber, Georg, Schrödel, Silke, Thirion, Christian, and Stoiber, Heribert
- Subjects
COMPLEMENT factor H ,CHRONIC lymphocytic leukemia ,CYTOTOXINS ,KILLER cells ,CANCER cells - Abstract
Antibody-mediated complement-dependent cytotoxicity (CDC) on malignant cells is regulated by several complement control proteins, including the inhibitory complement factor H (fH). fH consists of 20 short consensus repeat elements (SCRs) with specific functional domains. Previous research revealed that the fH-derived SCRs 19-20 (SCR1920) can displace full-length fH on the surface of chronic lymphocytic leukemia (CLL) cells, which sensitizes CLL cells for e.g. CD20-targeting therapeutic monoclonal antibody (mAb) induced CDC. Therefore, we constructed lentiviral vectors for the generation of cell lines that stably produce mAb-SCR-fusion variants starting from the clinically approved parental mAbs rituximab, obinutuzumab and ofatumumab, respectively. Flowcytometry revealed that the modification of the mAbs by the SCRs does not impair the binding to CD20. Increased in vitro lysis potency compared to their parental mAbs was corroborated by showing specific and dose dependent target cell elimination by CDC when compared to their parental mAbs. Lysis of CLL cells was not affected by the depletion of NK cells, suggesting that antibodydependent cellular cytotoxicity plays a minor role in this context. Overall, this study emphasizes the crucial role of CDC in the elimination of CLL cells by mAbs and introduces a novel approach for enhancing CDC by directly fusing fH SCR1920 with mAbs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Genetic factors associated with age-related macular degeneration modulating plasma inflammatory biomarker levels in patients with AIDS.
- Author
-
Sezgin, Efe, Schneider, Michael F., Hunt, Peter W., Beck-Engeser, Gabriele, Ambayac, Gabriele C., and Jabs, Douglas A.
- Subjects
- *
AIDS , *MACULAR degeneration , *AIDS complications , *TUMOR necrosis factor receptors , *COMPLEMENT factor H - Abstract
Introduction: Patients with the acquired immunodeficiency syndrome (AIDS) have an increased prevalence and incidence of intermediate-stage age-related macular degeneration (AMD). Several elevated plasma inflammatory biomarkers are associated with increased incidence of intermediate-stage AMD in this population. We evaluated the association between AMD risk alleles and plasma inflammatory biomarker levels in persons with AIDS. Materials and Methods: Cryopreserved plasma specimens of 229 non-Hispanic White and 252 non-Hispanic blacks from the Longitudinal Study of the Ocular Complications of AIDS cohort were assayed for plasma levels of soluble tumor necrosis factor receptor (sTNFR) 2, interleukin (IL)-18, C × 3motif chemokine ligand 1 (CX3CL1), C-reactive protein (CRP), and soluble CD14 (sCD14). Genotyping included AMD-associated variants rs10801553 and rs800292 for complement factor H (CFH) rs9332739 and rs547154 for complement factor 2 (C2), rs2230199 for C3, rs2285714 for CFI, and rs3732379 and rs3732378 for C × 3motif chemokine receptor 1 (CX3CR1). Results: In Whites, AMD low-risk CX3CR1 variants (V249I and T280M) were associated with reduced plasma levels of IL-18. In Blacks, AMD low-risk C3 R102G and low-risk CX3CR1 T280M variants were associated with reduced CRP levels. Conclusions: Genetic variants in AMD-associated immune genes may influence AMD-associated systemic plasma inflammatory biomarker levels in patients with AIDS. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Idiopathic sudden sensorineural hearing loss: A review focused on the contribution of vascular pathologies.
- Author
-
Tsuzuki, Nobuyoshi and Wasano, Koichiro
- Subjects
- *
SENSORINEURAL hearing loss , *IDIOPATHIC diseases , *DYSLIPIDEMIA , *CARDIOVASCULAR diseases , *COMPLEMENT factor H , *PATHOLOGY , *HEARING disorders - Abstract
Idiopathic sudden sensorineural hearing loss (ISSNHL) is characterized by abruptly appearing hearing loss, sometimes accompanied by vertigo. Vascular pathologies (e.g., cochlear ischemia, or cochlear infarction) are one of the most likely causes of ISSNHL. This review aims to present current understanding of inner ear anatomy, clinical features of ISSNHL, and its treatment strategies. The labyrinthine artery is the only end artery supplying blood to the inner ear, and it has three branches: the anterior vestibular artery, the main cochlear artery, and the vestibulo-cochlear artery (VCA). Occlusion of the VCA can be caused by a variety of factors. The VCA courses through a narrow bone canal. ISSNHL is usually diagnosed after excluding retrocochlear pathologies of sudden sensorineural hearing loss (SSNHL), such as vestibular schwannoma. Therefore, a head MRI or assessing auditory brainstem responses are recommended for patients with SSNHL. Severe SSNHL patients with high CHADS 2 scores, an index of stroke risk, have a significantly lower rate of vestibular schwannoma than severe SSNHL patients with low CHADS 2 scores, suggesting that severe ISSNHL in individuals at high risk of stroke is caused by vascular impairments. Intralabyrinthine hemorrhage causes SSNHL or vertigo, as in ISSNHL. The diagnosis of intralabyrinthine hemorrhage requires careful interpretation of MRI, and a small percentage of patients diagnosed with ISSNHL may in fact have intralabyrinthine hemorrhage. Many studies have reported an association between ISSNHL and atherosclerosis or cardiovascular risk factors (e.g., diabetes mellitus, hypertension, dyslipidemia and cardiovascular disease), and subsequent risk of stroke in patients with ISSNHL may be elevated compared to controls. Increased hearing level on the healthy ear side, high Framingham risk score, high neutrophil-to-lymphocyte ratio, high platelet-to-lymphocyte ratio, and severe white matter lesions may be poor prognostic factors for patients with ISSNHL. The association between thrombosis-related genes and susceptibility to ISSNHL has been reported in many studies (e.g., coagulation factor 2, coagulation factor 5, plasminogen activator inhibitor-1, platelet-associated genes, a homocysteine metabolism-related enzyme gene, endothelin-1, nitric oxide 3, phosphodiesterase 4D, complement factor H, and protein kinase C-eta). Treatment of ISSNHL with the aim of mitigating the vascular impairment in the inner ear includes systemically administered steroids, intratympanic steroid injections, hyperbaric oxygen therapy, prostaglandin E1, defibrinogenation therapy, and hydrogen inhalation therapy, but there is currently no evidence-based treatment for ISSNHL. Breakthroughs in the unequivocal diagnosis and treatment of ISSNHL due to vascular impairment are crucial to improve quality of life. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Tolerability and Safety of HF1K16 Injection in Patients With Refractory Solid Tumors
- Author
-
Tigermed Consulting Co., Ltd
- Published
- 2024
13. Evaluation of Safety and Immunogenicity of Ad26.Mos4.HIV and CH505 TF chTrimer Combination in Healthy Adults (RV591)
- Author
-
Janssen Vaccines & Prevention B.V., Henry M. Jackson Foundation for the Advancement of Military Medicine, Duke University, Walter Reed Army Institute of Research (WRAIR), National Institute of Allergy and Infectious Diseases (NIAID), and Makerere University Walter Reed Project
- Published
- 2024
14. The AKT2/SIRT5/TFEB pathway as a potential therapeutic target in non-neovascular AMD.
- Author
-
Ghosh, Sayan, Sharma, Ruchi, Bammidi, Sridhar, Koontz, Victoria, Nemani, Mihir, Yazdankhah, Meysam, Kedziora, Katarzyna M., Stolz, Donna Beer, Wallace, Callen T., Yu-Wei, Cheng, Franks, Jonathan, Bose, Devika, Shang, Peng, Ambrosino, Helena M., Dutton, James R., Geng, Zhaohui, Montford, Jair, Ryu, Jiwon, Rajasundaram, Dhivyaa, and Hose, Stacey
- Subjects
MACULAR degeneration ,COMPLEMENT factor H ,TRANSCRIPTION factors ,LYSOSOMES ,MITOCHONDRIA - Abstract
Non-neovascular or dry age-related macular degeneration (AMD) is a multi-factorial disease with degeneration of the aging retinal-pigmented epithelium (RPE). Lysosomes play a crucial role in RPE health via phagocytosis and autophagy, which are regulated by transcription factor EB/E3 (TFEB/E3). Here, we find that increased AKT2 inhibits PGC-1α to downregulate SIRT5, which we identify as an AKT2 binding partner. Crosstalk between SIRT5 and AKT2 facilitates TFEB-dependent lysosomal function in the RPE. AKT2/SIRT5/TFEB pathway inhibition in the RPE induced lysosome/autophagy signaling abnormalities, disrupted mitochondrial function and induced release of debris contributing to drusen. Accordingly, AKT2 overexpression in the RPE caused a dry AMD-like phenotype in aging Akt2 KI mice, as evident from decline in retinal function. Importantly, we show that induced pluripotent stem cell-derived RPE encoding the major risk variant associated with AMD (complement factor H; CFH Y402H) express increased AKT2, impairing TFEB/TFE3-dependent lysosomal function. Collectively, these findings suggest that targeting the AKT2/SIRT5/TFEB pathway may be an effective therapy to delay the progression of dry AMD. Lysosomes maintain RPE health via TFEB/E3-regulated autophagy. Here, the authors show deregulated AKT2/SIRT5/TFEB signaling in the RPE inhibits both lysosomal and mitochondrial function and leads to AMD, suggesting this pathway might provide a therapeutic target for AMD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Anti-C5 monoclonal antibody treatment showing pathological resolution of complement-mediated atypical hemolytic uremic syndrome: a case report.
- Author
-
Kurihara, Shigekazu, Yamaguchi, Akinori, Sonoda, Kosuke, Yamada, Yosuke, Harada, Makoto, Hashimoto, Koji, Shimojo, Hisashi, Ikeda, Yoichiro, and Kamijo, Yuji
- Subjects
HEMOLYTIC-uremic syndrome ,COMPLEMENT factor H ,MONOCLONAL antibodies ,THROMBOTIC thrombocytopenic purpura ,HEALTH facilities ,RENAL biopsy - Abstract
Background: No reports have shown histological changes before and after anti-C5 monoclonal antibody treatment in patients with atypical hemolytic uremic syndrome (aHUS). Here, we report a rare case of complement-mediated aHUS with a complement factor H (CFH) mutation and anti-CFH antibodies who underwent multiple kidney biopsies. Case presentation: A 53-year-old woman developed aHUS with CFH gene mutation [c.3572C > T (p. Ser1191 Leu)] and anti-CFH antibodies. Her father had succumbed to acute kidney injury (AKI) in his 30 s. She exhibited AKI, thrombocytopenia, and hemolytic anemia with schistocytes. After improving the platelet count with one session of plasma exchange, a kidney biopsy was performed one month after the onset of symptoms. Blood vessel thrombosis, obvious endothelial swelling, endocapillary hypercellularity, and subendothelial exudative lesions in the glomeruli and arterioles were detected. Anti-C5 monoclonal antibody treatment with eculizumab immediately improved disease activity. A second biopsy 3 months later revealed marked improvement of endothelial injuries with residual membrane double contours and exudative lesions. A third biopsy at 17 months after gradual improvement of kidney function showed a further decrease of double contours along with alterations of the exudative lesions to fibrous intimal thickening. Conclusions: This is the first report showing the pathophysiology of aHUS in the kidneys and the efficacy of anti-C5 monoclonal antibody treatment by presenting serial kidney pathological features before and after anti-C5 monoclonal antibody treatment. Since her CFH mutation was considered the most important pathological condition, treatment centered on eculizumab was administered, resulting in a good long-term prognosis. In addition, kidney pathological resolution in aHUS occurred over 1 year after anti-C5 monoclonal antibody treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Genetic Insights into Age-Related Macular Degeneration.
- Author
-
Bhumika, Bora, Nalini S., and Bora, Puran S.
- Subjects
MACULAR degeneration ,EXTRACELLULAR matrix proteins ,COMPLEMENT factor H ,VISION disorders ,GENETIC variation - Abstract
One of the major causes of vision impairment among elderly people in developed nations is age-related macular degeneration (AMD). The distinctive features of AMD are the accumulation of extracellular deposits called drusen and the gradual deterioration of photoreceptors and nearby tissues in the macula. AMD is a complex and multifaceted disease influenced by several factors such as aging, environmental risk factors, and a person's genetic susceptibility to the condition. The interaction among these factors leads to the initiation and advancement of AMD, where genetic predisposition plays a crucial role. With the advent of high-throughput genotyping technologies, many novel genetic loci associated with AMD have been identified, enhancing our knowledge of its genetic architecture. The common genetic variants linked to AMD are found on chromosome 1q32 (in the complement factor H gene) and 10q26 (age-related maculopathy susceptibility 2 and high-temperature requirement A serine peptidase 1 genes) loci, along with several other risk variants. This review summarizes the common genetic variants of complement pathways, lipid metabolism, and extracellular matrix proteins associated with AMD risk, highlighting the intricate pathways contributing to AMD pathogenesis. Knowledge of the genetic underpinnings of AMD will allow for the future development of personalized diagnostics and targeted therapeutic interventions, paving the way for more effective management of AMD and improved outcomes for affected individuals. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Proof of concept of a new plasma complement Factor H from waste plasma fraction.
- Author
-
Mori, Filippo, Pascali, Giancarlo, Berra, Silvia, Lazzarotti, Alessandra, Panetta, Daniele, Rocchiccioli, Silvia, Ceccherini, Elisa, Norelli, Francesco, Morlando, Antonio, Donadelli, Roberta, Clivio, Alberto, Farina, Claudio, Noris, Marina, Salvadori, Piero A., and Remuzzi, Giuseppe
- Subjects
COMPLEMENT factor H ,PROOF of concept ,POSITRON emission tomography ,BLOOD plasma ,ANIMAL experimentation - Abstract
Introduction: Complement factor H (FH) is a major regulator of the complement alternative pathway, its mutations predispose to an uncontrolled activation in the kidney and on blood cells and to secondary C3 deficiency. Plasma exchange has been used to correct for FH deficiency and although the therapeutic potential of purified FH has been suggested by in vivo experiments in animal models, a clinical approved FH concentrate is not yet available. We aimed to develop a purification process of FH from a waste fraction rather than whole plasma allowing a more efficient and ethical use of blood and plasma donations. Methods: Waste fractions from industrial plasma fractionation (pooled human plasma) were analyzed for FH content by ELISA. FH was purified from unused fraction III and its decay acceleration, cofactor, and C3 binding capacity were characterized in vitro. Biodistribution was assessed by high-resolution dynamic PET imaging. Finally, the efficacy of the purified FH preparation was tested in the mouse model of C3 glomerulopathy (Cfh-/- mice). Results: Our purification method resulted in a high yield of highly purified (92,07%), pathogen-safe FH. FH concentrate is intact and fully functional as demonstrated by in vitro functional assays. The biodistribution revealed lower renal and liver clearance of human FH in Cfh-/- mice than in wt mice. Treatment of Cfh-/- mice documented its efficacy in limiting C3 activation and promoting the clearance of C3 glomerular deposits. Conclusion: We developed an efficient and economical system for purifying intact and functional FH, starting from waste material of industrial plasma fractionation. The FH concentrate could therefore constitute possible treatments options of patients with C3 glomerulopathy, particularly for those with FH deficiency, but also for patients with other diseases associated with alternative pathway activation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Role of Complement Components in Asthma: A Systematic Review.
- Author
-
Tornyi, Ilona and Horváth, Ildikó
- Subjects
- *
COMPLEMENT (Immunology) , *COMPLEMENT factor H , *ASTHMA , *ASTHMATICS , *COMPLEMENT activation , *WHEEZE - Abstract
Background: Asthma is a chronic inflammatory airway disease characterized by recurrent symptoms in response to a wide range of external stimuli, including allergens, viral infections, and air pollution together with internal host-derived danger signals. The disease is traditionally associated with adaptive immune responses; recent research emphasizes the critical role of innate immunity in its pathogenesis. The complement system, activated as part of the defense mechanisms, plays a crucial role in bridging innate to adaptive immunity. While experimental models demonstrate complement cascade activation in asthma, human studies remain limited. Methods: This systematic review summarizes existing literature on the complement system in asthma patients, gathering data from PubMed, Web of Science, Scopus, and Google Scholar. The protocol was registered in the OSF. Results: Out of 482 initially identified articles, only 24 met the eligibility criteria, revealing disparities in sample origin, methodologies, and populations. Despite observed heterogeneity, a consistent result was found in the elevation of complement regulatory proteins, such as complement Factor H, in samples from patients with asthma compared to those from healthy subjects. Conclusions: The increased level of regulatory proteins, such as Factor H and I highlight that these may influence asthma pathophysiology. The role of complement factors as potential biomarkers of asthma activity and severity needs further evaluation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. Targeted complement inhibition using bispecific antibodies that bind local antigens and endogenous complement regulators.
- Author
-
Haiyu Wang, van de Bovenkamp, Fleur S., Dijkstra, Douwe J., Abendstein, Leoni, Borggreven, Nicole V., Pool, Jos, Zuijderduijn, Rob, Gstöttner, Christoph, Gelderman, Kyra A., Damelang, Timon, Vidarsson, Gestur, Blom, Anna M., Domínguez-Vega, Elena, Parren, Paul W. H. I., Sharp, Thomas H., and Trouw, Leendert A.
- Subjects
COMPLEMENT inhibition ,BISPECIFIC antibodies ,ECULIZUMAB ,COMPLEMENT factor H ,CELL surface antigens ,COMPLEMENT activation - Abstract
Complement activation protects against infection but also contributes to pathological mechanisms in a range of clinical conditions such as autoimmune diseases and transplant rejection. Complement-inhibitory drugs, either approved or in development, usually act systemically, thereby increasing the risk for infections. We therefore envisioned a novel class of bispecific antibodies (bsAbs) which are capable of site-directed complement inhibition by bringing endogenous complement regulators in the vicinity of defined cell surface antigens. Here, we analyzed a comprehensive set of obligate bsAbs designed to crosslink a specific target with either complement regulator factor H (FH) or C4b-binding protein (C4BP). The bsAbs were assessed for their capacity to inhibit complement activation and cell lysis in an antigen-targeted manner. We observed that the bsAbs inhibited classical, lectin, and alternative pathway complement activation in which sufficient endogenous serum FH and C4BP could be recruited to achieve local inhibition. Importantly, the bsAbs effectively protected antigen-positive liposomes, erythrocytes, and human leukocytes from complement-mediated lysis. In conclusion, localized complement inhibition by bsAbs capable of recruiting endogenous human complement regulators (such as FH or C4BP) to cell surfaces potentially provides a novel therapeutic approach for the targeted treatment of complement-mediated diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. Moss-produced human complement factor H with modified glycans has an extended half-life and improved biological activity.
- Author
-
Tschongov, Todor, Konwar, Swagata, Busch, Andreas, Sievert, Christian, Hartmann, Andrea, Noris, Marina, Gastoldi, Sara, Aiello, Sistiana, Schaaf, Andreas, Panse, Jens, Zipfel, Peter F., Dabrowska-Schlepp, Paulina, and Häffner, Karsten
- Subjects
COMPLEMENT factor H ,HEMOLYTIC-uremic syndrome ,GLYCANS ,COMPLEMENT activation ,INDUSTRIAL costs - Abstract
Most drugs that target the complement system are designed to inhibit the complement pathway at either the proximal or terminal levels. The use of a natural complement regulator such as factor H (FH) could provide a superior treatment option by restoring the balance of an overactive complement system while preserving its normal physiological functions. Until now, the systemic treatment of complement-associated disorders with FH has been deemed unfeasible, primarily due to high production costs, risks related to FH purified from donors' blood, and the challenging expression of recombinant FH in different host systems. We recently demonstrated that a moss-based expression system can produce high yields of properly folded, fully functional, recombinant FH. However, the half-life of the initial variant (CPV-101) was relatively short. Here we show that the same polypeptide with modified glycosylation (CPV-104) achieves a pharmacokinetic profile comparable to that of native FH derived from human serum. The treatment of FH-deficient mice with CPV-104 significantly improved important efficacy parameters such as the normalization of serum C3 levels and the rapid degradation of C3 deposits in the kidney compared to treatment with CPV-101. Furthermore, CPV-104 showed comparable functionality to serum-derived FH in vitro, as well as similar performance in ex vivo assays involving samples from patients with atypical hemolytic uremic syndrome, C3 glomerulopathy and paroxysomal nocturnal hematuria. CPV-104 - the human FH analog expressed in moss - will therefore allow the treatment of complement-associated human diseases by rebalancing instead of inhibiting the complement cascade. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. In vitro suppression of porcine epidemic diarrhea virus by Panax notoginseng saponins: assessing antiviral potential.
- Author
-
Hu, Yiyi, Li, Yunchuan, Zhu, Haodan, Wang, Dandan, Zhou, Junming, Ni, Yanxiu, Guo, Rongli, Fan, Baochao, and Li, Bin
- Subjects
- *
PORCINE epidemic diarrhea virus , *CADHERINS , *INTERFERON receptors , *TYPE I interferons , *INTERLEUKIN receptors , *COMPLEMENT factor H , *SAPONINS , *G protein coupled receptors , *PANAX - Abstract
Porcine epidemic diarrhea virus (PEDV) causes severe diarrhea and high mortality in neonatal suckling piglets, leading to significant economic losses to the swine industry. Panax notoginseng saponins (PNS) are bioactive extracts derived from the P. notoginseng plant. In this study, we investigated the anti-PEDV effect of PNS by employing various methodologies to assess their impact on PEDV in Vero cells. Using a CCK-8 (Cell Counting Kit-8) assay, we found that PNS had no significant cytotoxicity below the concentration of 128 µg/mL in Vero cells. Using immunofluorescence assays (IFAs), an enzyme-linked immunosorbent assay (ELISA), and plaque formation assays, we observed a dose-dependent inhibition of PEDV infection by PNS within 24–48 hours postinfection. PNS exerts its anti-PEDV activity specifically at the genome replication stage, and mRNA-seq analysis demonstrated that treatment with PNS resulted in increased expression of various genes, including IFIT1 (interferon-induced protein with tetratricopeptide repeats 1), IFIT3 (interferon-induced protein with tetratricopeptide repeats 3), CFH (complement factor H), IGSF10 (immunoglobulin superfamily member 10), ID2 (inhibitor of DNA binding 2), SPP1 (secreted phosphoprotein 1), PLCB4 (phospholipase C beta 4), and FABP4 (fatty acid binding protein 4), but it resulted in decreased expression of IL1A (interleukin 1 alpha), TNFRSF19 (TNF receptor superfamily member 19), CDH8 (cadherin 8), DDIT3 (DNA damage inducible transcript 3), GADD45A (growth arrest and DNA damage inducible alpha), PTPRG (protein tyrosine phosphatase receptor type G), PCK2 (phosphoenolpyruvate carboxykinase 2), and ADGRA2 (adhesion G protein-coupled receptor A2). This study provides insights into the potential mechanisms underlying the antiviral effects of PNS. Taken together, the results suggest that the PNS might effectively regulate the defense response to the virus and have potential to be used in antiviral therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Addressing the impact of high glucose microenvironment on the immunosuppressive characteristics of human mesenchymal stem cells.
- Author
-
Khasawneh, Ramada R., Abu‐El‐Rub, Ejlal, Almahasneh, Fatimah A., Alzu'bi, Ayman, Zegallai, Hana M., Almazari, Rawan A., Magableh, Huthaifa, Mazari, Mohammad H., Shlool, Haitham F., and Sanajleh, Ahmad K.
- Subjects
- *
MESENCHYMAL stem cells , *HUMAN stem cells , *COMPLEMENT factor H , *HYPERGLYCEMIA , *GLUCOSE , *CELL physiology - Abstract
Mesenchymal stem cells (MSCs) are a therapeutically efficient type of stem cells validated by their ability to treat many inflammatory and chronic conditions. The biological and therapeutic characteristics of MSCs can be modified depending on the type of microenvironment at the site of transplantation. Diabetes mellitus (DM) is a commonly diagnosed metabolic disease characterized by hyperglycemia, which alters over time the cellular and molecular functions of many cells and causes their damage. Hyperglycemia can also impact the success rate of MSCs transplantation; therefore, it is extremely significant to investigate the effect of high glucose on the biological and therapeutic attributes of MSCs, particularly their immunomodulatory abilities. Thus, in this study, we explored the effect of high glucose on the immunosuppressive characteristics of human adipose tissue‐derived mesenchymal stem cells (hAD‐MSCs). We found that hAD‐MSCs cultured in high glucose lost their immunomodulatory abilities and became detectable by immune cells. The decline in the immunosuppressive capabilities of hAD‐MSCs was mediated by significant decrease in the levels of IDO, IL‐10, and complement factor H and substantial increase in the activity of immunoproteasome. The protein levels of AMP‐activated protein kinase (AMPK) and phosphofructokinase‐1 (PFK‐1), which are integral regulators of glycolysis, revealed a marked decline in high glucose exposed MSCs. The findings of our study indicated the possibility of immunomodulatory shift in MSCs after being cultured in high glucose, which can be translationally employed to explain their poor survival and short‐lived therapeutic outcomes in diabetic patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. Corrigendum: Genetic investigation of Nordic patients with complement-mediated kidney diseases.
- Subjects
COMPLEMENT factor H ,BONE morphogenetic proteins ,MACULAR degeneration ,HEMOLYTIC-uremic syndrome ,COMPLEMENT (Immunology) - Abstract
This document is a corrigendum published in the journal Frontiers in Immunology. It corrects errors in a previously published article titled "Genetic investigation of Nordic patients with complement-mediated kidney diseases." The errors include mistakes in Table 3 and Supplementary Table 1, which have now been corrected. The authors apologize for the errors but state that they do not affect the scientific conclusions of the article. The corrigendum also includes information about the publisher's note and copyright information. [Extracted from the article]
- Published
- 2024
- Full Text
- View/download PDF
24. Eculizumab as first-line treatment for patients with severe presentation of complement factor H antibody–mediated hemolytic uremic syndrome
- Author
-
Coccia, Paula A., Alconcher, Laura F., Ferraris, Veronica, Lucarelli, Lucas I., Grillo, Maria A., Arias, Maria Andrea, Saurit, Mariana, Ratto, Viviana M., dos Santos, Celia, and Sánchez-Luceros, Analía
- Published
- 2024
- Full Text
- View/download PDF
25. Systems genomics in age-related macular degeneration
- Author
-
Hollander, Anneke I den, Mullins, Robert F, Orozco, Luz D, Voigt, Andrew P, Chen, Hsu-Hsin, Strunz, Tobias, Grassmann, Felix, Haines, Jonathan L, Kuiper, Jonas JW, Tumminia, Santa J, Allikmets, Rando, Hageman, Gregory S, Stambolian, Dwight, Klaver, Caroline CW, Boeke, Jef D, Chen, Hao, Honigberg, Lee, Katti, Suresh, Frazer, Kelly A, Weber, Bernhard HF, and Gorin, Michael B
- Subjects
Biomedical and Clinical Sciences ,Ophthalmology and Optometry ,Macular Degeneration ,Neurosciences ,Eye Disease and Disorders of Vision ,Human Genome ,Stem Cell Research ,Neurodegenerative ,Biotechnology ,Genetics ,Stem Cell Research - Induced Pluripotent Stem Cell ,Aging ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,2.1 Biological and endogenous factors ,Aetiology ,Eye ,Humans ,Retinal Pigment Epithelium ,Complement System Proteins ,Choroid ,Proteins ,Genomics ,Polymorphism ,Single Nucleotide ,Complement Factor H ,High-Temperature Requirement A Serine Peptidase 1 ,Age-related macular degeneration ,Omics ,Systems genomics ,Single cell sequencing ,Expression quantitative trait locus ,Complement system ,iPSc-RPE ,Induced pluripotent stem cells ,Clinical trial ,Polygenic risk scores ,Medical Biochemistry and Metabolomics ,Opthalmology and Optometry ,Ophthalmology & Optometry ,Ophthalmology and optometry - Abstract
Genomic studies in age-related macular degeneration (AMD) have identified genetic variants that account for the majority of AMD risk. An important next step is to understand the functional consequences and downstream effects of the identified AMD-associated genetic variants. Instrumental for this next step are 'omics' technologies, which enable high-throughput characterization and quantification of biological molecules, and subsequent integration of genomics with these omics datasets, a field referred to as systems genomics. Single cell sequencing studies of the retina and choroid demonstrated that the majority of candidate AMD genes identified through genomic studies are expressed in non-neuronal cells, such as the retinal pigment epithelium (RPE), glia, myeloid and choroidal cells, highlighting that many different retinal and choroidal cell types contribute to the pathogenesis of AMD. Expression quantitative trait locus (eQTL) studies in retinal tissue have identified putative causal genes by demonstrating a genetic overlap between gene regulation and AMD risk. Linking genetic data to complement measurements in the systemic circulation has aided in understanding the effect of AMD-associated genetic variants in the complement system, and supports that protein QTL (pQTL) studies in plasma or serum samples may aid in understanding the effect of genetic variants and pinpointing causal genes in AMD. A recent epigenomic study fine-mapped AMD causal variants by determing regulatory regions in RPE cells differentiated from induced pluripotent stem cells (iPSC-RPE). Another approach that is being employed to pinpoint causal AMD genes is to produce synthetic DNA assemblons representing risk and protective haplotypes, which are then delivered to cellular or animal model systems. Pinpointing causal genes and understanding disease mechanisms is crucial for the next step towards clinical translation. Clinical trials targeting proteins encoded by the AMD-associated genomic loci C3, CFB, CFI, CFH, and ARMS2/HTRA1 are currently ongoing, and a phase III clinical trial for C3 inhibition recently showed a modest reduction of lesion growth in geographic atrophy. The EYERISK consortium recently developed a genetic test for AMD that allows genotyping of common and rare variants in AMD-associated genes. Polygenic risk scores (PRS) were applied to quantify AMD genetic risk, and may aid in predicting AMD progression. In conclusion, genomic studies represent a turning point in our exploration of AMD. The results of those studies now serve as a driving force for several clinical trials. Expanding to omics and systems genomics will further decipher function and causality from the associations that have been reported, and will enable the development of therapies that will lessen the burden of AMD.
- Published
- 2022
26. C-reactive protein-complement factor H axis as a biomarker of activity in early and intermediate age-related macular degeneration.
- Author
-
Giralt, Lena, Figueras-Roca, Marc, De Luis Eguileor, Beatriz, Romero, Barbara, Zarranz-Ventura, Javier, Alforja, Socorro, Santiago, Francisca, Bolaños, Jennifer, Lozano, Francisco, Dotti-Boada, Marina, Sala-Puigdollers, Anna, Dura, Paula, Izquierdo-Serra, Jordi, Valero, Oliver, Adan, Alfredo, Fonollosa, Alex, and Molins, Blanca
- Subjects
MACULAR degeneration ,COMPLEMENT factor H ,C-reactive protein ,CHOROID ,BIOMARKERS - Abstract
Purpose: To determine and compare the serum levels of complement Factor H (FH), monomeric C-Reactive Protein (mCRP) and pentameric C-Reactive protein (pCRP) in patients with age-related macular degeneration (AMD) and to correlate them with clinical, structural and functional parameters. Methods: Cross-sectional observational study. One hundred thirty-nine individuals (88 patients and 51 healthy controls) from two referral centers were included and classified into three groups: early or intermediate AMD (n=33), advanced AMD (n=55), and age and sex matched healthy controls (n=51). Serum levels of FH, mCRP, and pCRP were determined and correlated with clinical and imaging parameters. Results: Patients with intermediate AMD presented FH levels significantly lower than controls [186.5 (72.1-931.8) µg/mL vs 415.2 (106.1-1962.2) µg/mL; p=0.039] and FH levels <200 µg/mL were associated with the presence of drusen and pigmentary changes in the fundoscopy (p=0.002). While no differences were observed in pCRP and mCRP levels, and mCRP was only detected in less than 15% of the included participants, women had a significantly higher detection rate of mCRP than men (21.0% vs. 3.8%, p=0.045). In addition, the ratio mCRP/FH (log) was significantly lower in the control group compared to intermediate AMD (p=0.031). Visual acuity (p<0.001), macular volume (p<0.001), and foveal thickness (p=0.034) were significantly lower in the advanced AMD group, and choroidal thickness was significantly lower in advanced AMD compared to early/intermediate AMD (p=0.023). Conclusion: Intermediate AMD was associated in our cohort with decreased serum FH levels together with increased serum mCRP/FH ratio. All these objective serum biomarkers may suggest an underlying systemic inflammatory process in early/intermediate AMD patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. PolySialic Acid Nanoparticles Actuate Complement-Factor-H-Mediated Inhibition of the Alternative Complement Pathway: A Safer Potential Therapy for Age-Related Macular Degeneration.
- Author
-
Peterson, Sheri L., Krishnan, Anitha, Patel, Diyan, Khanehzar, Ali, Lad, Amit, Shaughnessy, Jutamas, Ram, Sanjay, Callanan, David, Kunimoto, Derek, Genead, Mohamed A., and Tolentino, Michael J.
- Subjects
- *
MACULAR degeneration , *COMPLEMENT inhibition , *COMPLEMENT factor H , *OPTIC neuritis , *COMPLEMENT activation , *RETROLENTAL fibroplasia - Abstract
The alternative pathway of the complement system is implicated in the etiology of age-related macular degeneration (AMD). Complement depletion with pegcetacoplan and avacincaptad pegol are FDA-approved treatments for geographic atrophy in AMD that, while effective, have clinically observed risks of choroidal neovascular (CNV) conversion, optic neuritis, and retinal vasculitis, leaving room for other equally efficacious but safer therapeutics, including Poly Sialic acid (PSA) nanoparticle (PolySia-NP)-actuated complement factor H (CFH) alternative pathway inhibition. Our previous paper demonstrated that PolySia-NP inhibits pro-inflammatory polarization and cytokine release. Here, we extend these findings by investigating the therapeutic potential of PolySia-NP to attenuate the alternative complement pathway. First, we show that PolySia-NP binds CFH and enhances affinity to C3b. Next, we demonstrate that PolySia-NP treatment of human serum suppresses alternative pathway hemolytic activity and C3b deposition. Further, we show that treating human macrophages with PolySia-NP is non-toxic and reduces markers of complement activity. Finally, we describe PolySia-NP-treatment-induced decreases in neovascularization and inflammatory response in a laser-induced CNV mouse model of neovascular AMD. In conclusion, PolySia-NP suppresses alternative pathway complement activity in human serum, human macrophage, and mouse CNV without increasing neovascularization. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. Therapy‐induced senescent cancer cells exhibit complement activation and increased complement regulatory protein expression.
- Author
-
Abu‐Humaidan, Anas HA, Ismail, Mohammad A, Ahmad, Fatima M, Al Shboul, Sofian, Barham, Raghad, Tadros, Joud S, Alhesa, Ahmad, El‐Sadoni, Mohammed, Alotaibi, Moureq R, Ababneh, Nidaa A, and Saleh, Tareq
- Subjects
- *
COMPLEMENT activation , *PROTEIN expression , *BREAST , *COMPLEMENT factor H , *COMPLEMENT receptors , *CANCER cells , *NEOADJUVANT chemotherapy - Abstract
Therapy‐induced senescence (TIS) is a primary response to chemotherapy, contributing to untoward treatment outcomes such as evasion of immunosurveillance. Despite the established role of the complement system in the immune response to cancer, the role of complement in mediating the immune response against senescent tumor cells remains poorly understood. To explore this relationship, we exposed lung adenocarcinoma (A549), breast adenocarcinoma (MCF7) and pancreatic carcinoma (Panc‐1) cell lines to sublethal doses of either etoposide or doxorubicin to trigger TIS. Identification of TIS was based on morphological changes, upregulation of the senescence‐associated β‐galactosidase, p21Cip1 induction and lamin B1 downregulation. Using immunofluorescence microscopy, quantitative PCR, ELISA of conditioned media and in silico analysis, we investigated complement activation, complement protein expression, C3 levels in the conditioned media of senescent cells and secreted complement proteins as part of the senescence‐associated secretory phenotype (SASP), respectively. In cell lines undergoing TIS, complement‐related changes included (i) activation of the terminal pathway, evidenced by the deposition of C5b‐9 on senescent cells; (ii) an increase in the expression of CD59 and complement factor H and (iii) in A549 cells, an elevation in the expression of C3 with its secretion into the medium. In addition, increased C3 expression was observed in breast cancer samples expressing TIS hallmarks following exposure to neoadjuvant chemotherapy. In conclusion, TIS led to the activation of complement, upregulation of complement regulatory proteins and increased C3 expression. Complement appears to play a role in shaping the cancer microenvironment upon senescence induction. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. Efficacy of GalNAc C3 siRNAs in factor H-deficient mice with C3 glomerulopathy.
- Author
-
Zanchi, Cristina, Locatelli, Monica, Cerullo, Domenico, Aumiller, Verena, Corna, Daniela, Rottoli, Daniela, Schubert, Steffen, Noris, Marina, Tomasoni, Susanna, Remuzzi, Giuseppe, Zoja, Carlamaria, and Benigni, Ariela
- Subjects
- *
MICE , *SMALL interfering RNA , *COMPLEMENT (Immunology) , *COMPLEMENT factor H - Abstract
Complement alternative pathway (AP) dysregulation drives C3 glomerulopathy (C3G), a rare renal disorder characterized by glomerular C3 deposition and glomerular damage, for which no effective treatments are available. Blockade of complement C3 is emerging as a viable therapeutic option. In an earlier study we showed that SLN500, a small interfering RNA targeting liver C3 synthesis, was able to limit AP dysregulation and glomerular C3d deposits in mice with partial factor H (FH) deficiency (Cfh +/- mice). Here, we assessed the pharmacological effects of SLN501 – an optimized SLN500 version – in mice with complete FH deficiency (Cfh -/- mice) that exhibit a more severe C3G phenotype. SLN501 effectively prevented liver C3 synthesis, thus limiting AP dysregulation, glomerular C3d deposits and the development of ultrastructural alterations. These data provide firm evidence of the use of siRNA-mediated liver C3 gene silencing as a potential therapy for treating C3G patients with either partial or complete FH loss of function. • A GalNAc anti-C3 siRNA, SLN501, silences liver C3 synthesis in Cfh -/- mice. • SLN501 limits fluid-phase complement dysregulation in Cfh -/- mice. • SLN501 reduces glomerular C3d and mesangial electron-dense deposits in Cfh -/- mice. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. Triple-fusion protein (TriFu): A potent, targeted, enzyme-like inhibitor of all three complement activation pathways.
- Author
-
Sonnentag, Sophia J., Dopler, Arthur, Kleiner, Katharina, Garg, Brijesh K., Mannes, Marco, Späth, Nadja, Akilah, Amira, Höchsmann, Britta, Schrezenmeier, Hubert, Anliker, Markus, Boyanapalli, Ruby, Huber-Lang, Markus, and Schmidt, Christoph Q.
- Subjects
- *
COMPLEMENT activation , *COMPLEMENT factor H , *COMPLEMENT inhibition , *PAROXYSMAL hemoglobinuria , *SURFACE plasmon resonance , *COMPLEMENT receptors - Abstract
The introduction of a therapeutic anti-C5 antibody into clinical practice in 2007 inspired a surge into the development of complement-targeted therapies. This has led to the recent approval of a C3 inhibitory peptide, an antibody directed against C1s and a full pipeline of several complement inhibitors in preclinical and clinical development. However, no inhibitor is available that efficiently inhibits all three complement initiation pathways and targets host cell surface markers as well as complement opsonins. To overcome this, we engineered a novel fusion protein combining selected domains of the three natural complement regulatory proteins decay accelerating factor, factor H and complement receptor 1. Such a triple fusion complement inhibitor (TriFu) was recombinantly expressed and purified alongside multiple variants and its building blocks. We analyzed these proteins for ligand binding affinity and decay acceleration activity by surface plasmon resonance. Additionally, we tested complement inhibition in several in vitro/ex vivo assays using standard classical and alternative pathway restricted hemolysis assays next to hemolysis assays with paroxysmal nocturnal hemoglobinuria erythrocytes. A novel in vitro model of the alternative pathway disease C3 glomerulopathy was established to evaluate the potential of the inhibitors to stop C3 deposition on endothelial cells. Next to the novel engineered triple fusion variants which inactivate complement convertases in an enzyme-like fashion, stoichiometric complement inhibitors targeting C3, C5, factor B, and factor D were tested as comparators. The triple fusion approach yielded a potent complement inhibitor that effi- ciently inhibits all three complement initiation pathways while targeting to surface markers. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. Heterozygous mutations in factor H aggravate pathological damage in a stable IgA deposition model induced by Lactobacillus casei cell wall extract.
- Author
-
Jingyi Li, Yaping Dong, Feifei Chen, Hongyu Yang, Pei Chen, Hongyu Li, Sufang Shi, Xujie Zhou, Li Zhu, Yuemiao Zhang, Lijun Liu, Xinfang Xie, Feng Yu, Jing Jin, and Jicheng Lv
- Subjects
LACTOBACILLUS casei ,IMMUNOGLOBULIN A ,COMPLEMENT factor H ,IGA glomerulonephritis ,RENAL circulation - Abstract
Introduction: Activation of complement through the alternative pathway (AP) has a key role in the pathogenesis of IgA nephropathy (IgAN). We previously showed, by intraperitoneal injection of Lactobacillus casei cell wall extract (LCWE), C57BL/6 mice develop mild kidney damage in association with glomerular IgA deposition. To further address complement activity in causing glomerular histological alterations as suggested in the pathogenesis of IgAN, here we used mice with factor H mutation (FH
W/R ) to render AP overactivation in conjunction with LCWE injection to stimulate intestinal production of IgA. Methods: Dose response to LCWE were examined between two groups of FHW/R mice. Wild type (FHW/W) mice stimulated with LCWE were used as model control. Results: The FHW/R mice primed with high dose LCWE showed elevated IgA and IgA-IgG complex levels in serum. In addition to 100% positive rate of IgA and C3, they display elevated biomarkers of kidney dysfunction, coincided with severe pathological lesions, resembling those of IgAN. As compared to wild type controls stimulated by the same high dose LCWE, these FHW/R mice exhibited stronger complement activation in the kidney and in circulation. Discussion: The new mouse model shares many disease features with IgAN. The severity of glomerular lesions and the decline of kidney functions are further aggravated through complement overactivation. The model may be a useful tool for preclinical evaluation of treatment response to complement-inhibitors. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
32. Case report: A family of atypical hemolytic uremic syndrome involving a CFH:: CFHR1 fusion gene and CFHR3-1-4-2 gene duplication.
- Author
-
Yuko Tasaki, Hiroshi Tsujimoto, Tadafumi Yokoyama, Naotoshi Sugimoto, Shinji Kitajima, Hiroshi Fujii, Yoshihiko Hidaka, Noritoshi Kato, Shoichi Maruyama, Norimitsu Inoue, and Taizo Wada
- Subjects
HEMOLYTIC-uremic syndrome ,GENE fusion ,CHROMOSOME duplication ,COMPLEMENT factor H ,THROMBOTIC thrombocytopenic purpura - Abstract
Mutations in the complement factor H (CFH) gene are associated with complement dysregulation and the development of atypical hemolytic uremic syndrome (aHUS). Several fusion genes that result from genomic structural variation in the CFH and complement factor H-related (CFHR) gene regions have been identified in aHUS. However, one allele has both CFHR gene duplication and CFH::CFHR1 fusion gene have not been reported. An 8-month-old girl (proband) presented with aHUS and was treated with ravulizumab. Her paternal grandfather developed aHUS previously and her paternal great grandmother presented with anti-neutrophil cytoplasmic antibody-associated vasculitis and thrombotic microangiopathy (TMA). However, the proband's parents have no history of TMA. A genetic analysis revealed the presence of CFH::CFHR1 fusion gene and a CFHR3-1-4-2 gene duplication in the patient, her father, and her paternal grandfather. Although several fusion genes resulting from structural variations of the CFH-CFHR genes region have been identified, this is the first report of the combination of a CFH:: CFHR1 fusion gene with CFHR gene duplication. Because the CFH-CFHR region is highly homologous, we hypothesized that CFHR gene duplication occurred. These findings indicate a novel pathogenic genomic structural variation associated with the development of aHUS. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. Protective role of complement factor H against the development of preeclampsia.
- Author
-
Yasmin, Hadida, Agostinis, Chiara, Toffoli, Miriam, Roy, Tamali, Pegoraro, Silvia, Balduit, Andrea, Zito, Gabriella, Di Simone, Nicoletta, Ricci, Giuseppe, Madan, Taruna, Kishore, Uday, and Bulla, Roberta
- Subjects
COMPLEMENT factor H ,ECULIZUMAB ,PREECLAMPSIA ,COMPLEMENT inhibition ,PREGNANT women ,COMPLEMENT activation - Abstract
Pregnancy is an immunologically regulated, complex process. A tightly controlled complement system plays a crucial role in the successful establishment of pregnancy and parturition. Complement inhibitors at the feto-maternal interface are likely to prevent inappropriate complement activation to protect the fetus. In the present study, we aimed to understand the role of Factor H (FH), a negative regulator of complement activation, in normal pregnancy and in a model of pathological pregnancy, i.e. preeclampsia (PE). The distribution and expression of FH was investigated in placental tissues, various placental cells, and in the sera of healthy (CTRL) or PE pregnant women via immunohistochemistry, RT-qPCR, ELISA, and Western blot. Our results showed a differential expression of FH among the placental cell types, decidual stromal cells (DSCs), decidual endothelial cells (DECs), and extravillous trophoblasts (EVTs). Interestingly, FH was found to be considerably less expressed in the placental tissues of PE patients compared to normal placental tissue both at mRNA and protein levels. Similar results were obtained by measuring circulating FH levels in the sera of third trimester CTRL and PE mothers. Syncytiotrophoblast microvesicles, isolated from the placental tissues of PE and CTRL women, downregulated FH expression by DECs. The present study appears to suggest that FH is ubiquitously present in the normal placenta and plays a homeostatic role during pregnancy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. Thrombospondin-1, BIM and CFH polymorphisms and response to anti-VEGF treatment in neovascular age- related macular degeneration patients.
- Author
-
Sorenson, Christine M., Gurel, Zafer, Song, Yong-Seok, Peterson, Kyle D., Blodi, Barbara A., and Sheibani, Nader
- Subjects
- *
MACULAR degeneration , *COMPLEMENT factor H , *THROMBOSPONDIN-1 , *ENDOTHELIAL growth factors , *SINGLE nucleotide polymorphisms , *BEVACIZUMAB - Abstract
Age-related macular degeneration (AMD) is a vision threatening disease in older adults. Anti-VEGF treatment is effective for the majority of neovascular AMD (nAMD) patients, although approximately 30% of nAMD patients have an incomplete response for unknown reasons. Here we assessed the contribution of single nucleotide polymorphisms (SNPs) in key angioinflammatory regulatory genes in nAMD patients with an incomplete response compared to those responsive to anti-VEGF treatment. A total of 25 responsive and 30 nAMD patients with an incomplete response to anti-vascular endothelial growth factor (anti-VEGF) treatment were examined for known SNPs that impact the structure and function of thromobospondin-1 (TSP1), Bcl-2-interacting mediator of cell death (BIM) and complement factor H (CFH). Plasma levels of C-C motif chemokine ligand 2 (CCL2/MCP1), TSP1 and VEGF were assessed by ELISA. Patients responsive to anti-VEGF treatment showed a significant increase in the TSP1 rs2228262 AA allele and a trend for the BIM (rs724710) CT allele. Consistent with previous reports, 42% of the patients responsive to anti-VEGF expressed the CC allele for CFH rs1061170. Although the CFH TT allele had similarly low prevalence in both groups, the TC allele tended to be more prevalent in patients with an incomplete response. Patients with an incomplete response also had increased plasma CCL2/MCP1 levels, consistent with the role increased inflammation has in the pathogenesis of nAMD. Our studies point to new tools to assess the potential responsiveness of nAMD patients to anti-VEGF treatment and suggest the potential use of anti-CCL2 for treatment of nAMD patients with an incomplete response to anti-VEGF. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
35. The human factor H protein family -- an update.
- Author
-
Sándor, Noémi, Schneider, Andrea E., Matola, Alexandra T., Barbai, Veronika H., Bencze, Dániel, Hammad, Hani Hashim, Papp, Alexandra, Kövesdi, Dorottya, Uzonyi, Barbara, and Józsi, Mihály
- Subjects
ECULIZUMAB ,COMPLEMENT factor H ,COMPLEMENT activation ,MACULAR degeneration ,PROTEINS - Abstract
Complement is an ancient and complex network of the immune system and, as such, it plays vital physiological roles, but it is also involved in numerous pathological processes. The proper regulation of the complement system is important to allow its sufficient and targeted activity without deleterious side-effects. Factor H is a major complement regulator, and together with its splice variant factor H-like protein 1 and the five human factor H-related (FHR) proteins, they have been linked to various diseases. The role of factor H in inhibiting complement activation is well studied, but the function of the FHRs is less characterized. Current evidence supports the main role of the FHRs as enhancers of complement activation and opsonization, i.e., counter-balancing the inhibitory effect of factor H. FHRs emerge as soluble pattern recognition molecules and positive regulators of the complement system. In addition, factor H and some of the FHR proteins were shown to modulate the activity of immune cells, a non-canonical function outside the complement cascade. Recent efforts have intensified to study factor H and the FHRs and develop new tools for the distinction, quantification and functional characterization of members of this protein family. Here, we provide an update and overview on the versatile roles of factor H family proteins, what we know about their biological functions in healthy conditions and in diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. Complement factor H: a novel innate immune checkpoint in cancer immunotherapy.
- Author
-
Saxena, Ruchi, Gottlin, Elizabeth B., Campa, Michael J., Bushey, Ryan T., Jian Guo, Patz Jr., Edward F., and You-Wen He
- Subjects
COMPLEMENT factor H ,IMMUNE checkpoint proteins ,COMPLEMENT activation ,IMMUNOTHERAPY ,IMMUNE system - Abstract
The elimination of cancer cells critically depends on the immune system. However, cancers have evolved a variety of defense mechanisms to evade immune monitoring, leading to tumor progression. Complement factor H (CFH), predominately known for its function in inhibiting the alternative pathway of the complement system, has recently been identified as an important innate immunological checkpoint in cancer. CFH-mediated immunosuppression enhances tumor cells' ability to avoid immune recognition and produce an immunosuppressive tumor microenvironment. This review explores the molecular underpinnings, interactions with immune cells, clinical consequences, and therapeutic possibilities of CFH as an innate immune checkpoint in cancer control. The difficulties and opportunities of using CFH as a target in cancer immunotherapy are also explored. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
37. Factor H's Control of Complement Activation Emerges as a Significant and Promising Therapeutic Target for Alzheimer's Disease Treatment.
- Author
-
Hasantari, Iris, Nicolas, Nabil, Alzieu, Philippe, Leval, Léa, Shalabi, Andree, Grolleau, Sylvain, and Dinet, Virginie
- Subjects
- *
ALZHEIMER'S disease , *COMPLEMENT activation , *THERAPEUTICS , *COMPLEMENT factor H , *NEURAL transmission , *BLOOD-brain barrier - Abstract
The complement is a component of the innate immune system designed to fight infections and tissue- or age-related damages. Complement activation creates an inflammatory microenvironment, which enhances cell death. Excessive complement inflammatory activity has been linked to alterations in the structure and functions of the blood–brain barrier, contributing to a poor prognosis for Alzheimer's disease (AD). In the AD preclinical phase, individuals are often clinically asymptomatic despite evidence of AD neuropathology coupled with heightened inflammation. Considering the involvement of the complement system in the risk of developing AD, we hypothesize that inhibiting complement activation could reduce this inflammatory period observed even before clinical signs, thereby slowing down the onset/progression of AD. To validate our hypothesis, we injected complement inhibitor factor H into the brain of APP/PS1 AD mice at early or late stages of this pathology. Our results showed that the injection of factor H had effects on both the onset and progression of AD by reducing proinflammatory IL6, TNF-α, IL1β, MAC and amyloid beta levels. This reduction was associated with an increase in VGLUT1 and Psd95 synaptic transmission in the hippocampal region, leading to an improvement in cognitive functions. This study invites a reconsideration of factor H's therapeutic potential for AD treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. Enhancement of complement-dependent cytotoxicity by linking factor-H derived short consensus repeats 19-20 to CD20 antibodies
- Author
-
Lena Prantl, Philipp Heider, Lisa Bergmeister, Katharina Calana, Jan-Paul Bohn, Dominik Wolf, Zoltan Banki, Andreas Bosch, Maximilian Plach, Georg Huber, Silke Schrödel, Christian Thirion, and Heribert Stoiber
- Subjects
CLL ,complement-dependent cytotoxicity ,ofatumumab ,rituximab ,obinutuzumab ,complement factor H ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Antibody-mediated complement-dependent cytotoxicity (CDC) on malignant cells is regulated by several complement control proteins, including the inhibitory complement factor H (fH). fH consists of 20 short consensus repeat elements (SCRs) with specific functional domains. Previous research revealed that the fH-derived SCRs 19–20 (SCR1920) can displace full-length fH on the surface of chronic lymphocytic leukemia (CLL) cells, which sensitizes CLL cells for e.g. CD20-targeting therapeutic monoclonal antibody (mAb) induced CDC. Therefore, we constructed lentiviral vectors for the generation of cell lines that stably produce mAb-SCR-fusion variants starting from the clinically approved parental mAbs rituximab, obinutuzumab and ofatumumab, respectively. Flow-cytometry revealed that the modification of the mAbs by the SCRs does not impair the binding to CD20. Increased in vitro lysis potency compared to their parental mAbs was corroborated by showing specific and dose dependent target cell elimination by CDC when compared to their parental mAbs. Lysis of CLL cells was not affected by the depletion of NK cells, suggesting that antibody-dependent cellular cytotoxicity plays a minor role in this context. Overall, this study emphasizes the crucial role of CDC in the elimination of CLL cells by mAbs and introduces a novel approach for enhancing CDC by directly fusing fH SCR1920 with mAbs.
- Published
- 2024
- Full Text
- View/download PDF
39. A supernumerary synthetic chromosome in Komagataella phaffii as a repository for extraneous genetic material
- Author
-
Dariusz Abramczyk, Maria del Carmen Sanchez Olmos, Adan Andres Ramirez Rojas, Daniel Schindler, Daniel Robertson, Stephen McColm, Adele L. Marston, and Paul N. Barlow
- Subjects
Pichia pastoris ,Yeast ,Biomanufacture ,Artificial chromosome ,Therapeutic proteins ,Complement factor H ,Microbiology ,QR1-502 - Abstract
Abstract Background Komagataella phaffii (Pichia pastoris) is a methylotrophic commercially important non-conventional species of yeast that grows in a fermentor to exceptionally high densities on simple media and secretes recombinant proteins efficiently. Genetic engineering strategies are being explored in this organism to facilitate cost-effective biomanufacturing. Small, stable artificial chromosomes in K. phaffii could offer unique advantages by accommodating multiple integrations of extraneous genes and their promoters without accumulating perturbations of native chromosomes or exhausting the availability of selection markers. Results Here, we describe a linear “nano”chromosome (of 15–25 kb) that, according to whole-genome sequencing, persists in K. phaffii over many generations with a copy number per cell of one, provided non-homologous end joining is compromised (by KU70-knockout). The nanochromosome includes a copy of the centromere from K. phaffii chromosome 3, a K. phaffii-derived autonomously replicating sequence on either side of the centromere, and a pair of K. phaffii-like telomeres. It contains, within its q arm, a landing zone in which genes of interest alternate with long (approx. 1-kb) non-coding DNA chosen to facilitate homologous recombination and serve as spacers. The landing zone can be extended along the nanochromosome, in an inch-worming mode of sequential gene integrations, accompanied by recycling of just two antibiotic-resistance markers. The nanochromosome was used to express PDI, a gene encoding protein disulfide isomerase. Co-expression with PDI allowed the production, from a genomically integrated gene, of secreted murine complement factor H, a plasma protein containing 40 disulfide bonds. As further proof-of-principle, we co-expressed, from a nanochromosome, both PDI and a gene for GFP-tagged human complement factor H under the control of P AOX1 and demonstrated that the secreted protein was active as a regulator of the complement system. Conclusions We have added K. phaffii to the list of organisms that can produce human proteins from genes carried on a stable, linear, artificial chromosome. We envisage using nanochromosomes as repositories for numerous extraneous genes, allowing intensive engineering of K. phaffii without compromising its genome or weakening the resulting strain.
- Published
- 2023
- Full Text
- View/download PDF
40. Complement factor H attenuates TNF-α-induced inflammation by upregulating EIF3C in rheumatoid arthritis
- Author
-
Yimeng Jia, Bin Feng, Xin Ji, Xinping Tian, Lidan Zhao, Jiaxin Zhou, Wen Zhang, Mengtao Li, Yunyun Fei, and Xunyao Wu
- Subjects
Complement factor H ,Rheumatoid arthritis ,Pyroptosis ,Fibroblast-like synoviocytes ,Eukaryotic translation initiation factor 3 subunit C ,Medicine - Abstract
Abstract Objective To explore the role and underlying mechanism of Complement Factor H (CFH) in the peripheral and joint inflammation of RA patients. Methods The levels of CFH in the serum and synovial fluid were determined by ELISA. The pyroptosis of monocytes was determined by western blotting and flow cytometry. The inflammation cytokine release was tested by ELISA. The cell migration and invasion ability of fibroblast-like synoviocytes (FLS) were tested by Wound healing Assay and transwell assay, respectively. The potential target of CFH was identified by RNA sequencing. Results CFH levels were significantly elevated in the serum and synovial fluid from RA and associated with high sensitivity C-reactive protein (hs-CRP), erythrocyte sedimentation rate (ESR), and disease activity score 28 (DAS28). TNF-α could inhibit CFH expression, and CFH combined with TNF-α significantly decreased cell death, cleaved-caspase 3, gasdermin E N-terminal (GSDME-N), and inflammatory cytokines release (IL-1β and IL-6) of RA-derived monocytes. Stimulated with TNF-α increased CFH levels in RA FLS and CFH inhibits the migration, invasion, and TNF-α–induced production of inflammatory mediators, including proinflammatory cytokines (IL-6, IL-8) as well as matrix metalloproteinases (MMPs, MMP1 and MMP3) of RA FLSs. The RNA-seq results showed that CFH treatment induced upregulation of eukaryotic translation initiation factor 3 (EIF3C) in both RA monocytes and FLS. The migration of RA FLSs was promoted and the expressions of IL-6, IL-8, and MMP-3 were enhanced upon EIF3C knockdown under the stimulation of CFH combined with TNF-α. Conclusion In conclusion, we have unfolded the anti-inflammatory roles of CFH in the peripheral and joints of RA, which might provide a potential therapeutic target for RA patients.
- Published
- 2023
- Full Text
- View/download PDF
41. A Study to Assess the Effect of Food on the Pharmacokinetics of TY-9591 in Healthy Volunteers
- Published
- 2022
42. Therapies for Geographic Atrophy.
- Author
-
Gasparian, Suzie A., Skrehot, Henry C., and Weng, Christina Y.
- Subjects
- *
ATROPHY , *MACULAR degeneration , *POSTERIOR segment (Eye) , *COMPLEMENT factor H - Abstract
The article discusses therapies for geographic atrophy (GA), an advanced form of age-related macular degeneration (AMD) that causes progressive and irreversible blindness. The prevalence of AMD is increasing globally, with approximately 196 million people affected in 2020. Diagnosis is made through fundus examination and imaging techniques like fundus photography, fundus autofluorescence (FAF), and optical coherence tomography (OCT). Currently, there is no clear standard of care treatment for GA, and ophthalmologists rely on vitamins and visual rehabilitation to manage the condition. Several clinical trials investigating potential therapies have shown mixed results. One therapy, pegcetacoplan, has shown promising results in reducing GA growth rate in phase 2 and 3 trials. Another therapy, avacincaptad pegol, has also demonstrated significant reductions in GA growth rate. However, both therapies have been associated with an increased risk of developing choroidal neovascularization (CNV). Other investigational agents targeting oxidative stress, inflammation, and the complement pathway are also being studied. The most promising treatment option for GA is complement inhibition, which has shown to slow down the progression of the disease. Other therapies being pursued include gene therapy, regenerative and cell therapies. However, currently available treatments are unable to restore visual function and carry the risk of adverse events. Continued research and development of new drugs may lead to more effective treatments in the future. The document provides a list of references related to therapies for GA secondary [Extracted from the article]
- Published
- 2024
- Full Text
- View/download PDF
43. Discovery and validation of protein biomarkers for monitoring the effectiveness of drug treatment for major depressive disorder.
- Author
-
Lee, Seungyeon, Mun, Sora, Lee, Jiyeong, and Kang, Hee-Gyoo
- Subjects
- *
MENTAL depression , *DRUG efficacy , *DRUG monitoring , *HAMILTON Depression Inventory , *COMPLEMENT factor H - Abstract
Major depressive disorder (MDD) has a high prevalence worldwide. Although the economic burden of depression increases annually, the proportion of patients with MDD receiving treatment did not increase between 2010 and 2018, suggesting an unmet treatment need. The burden of long-term treatment for depression is borne by patients. In this context, biomarkers associated with drug-treatment responses can be used as reference indicators to reduce unnecessary treatment and costs. Changes in biomolecules in response to drug treatment for depression and drug-treatment response markers have been studied extensively. The Hamilton Depression Rating Scale (HAM-D) is mainly used as an indicator of response and remission; however, it is difficult to determine whether the medication contributes to recovery when evaluating the effect of drug treatment for depression based on this assessment. Therefore, it is necessary to monitor the effect of medication compared to normal health conditions. Here, serum protein levels were compared using liquid chromatography-tandem mass spectrometry among a group of patients with depression who did not receive medication, a group of patients receiving medication, and a control group. Eight selected biomarkers, including Apolipoproteins A-I, Complement factor H, Complement C5, Complement C1q subcomponent subunit B, Alpha-2-HS-glycoprotein, Complement C1q subcomponent subunit C, Vitamin D-binding protein and Corticosteroid-binding globulin were distinguished between disease states, and protein levels in the drug-treated group were similar to those in the control group. These markers can be used to monitor the effectiveness of drug treatment. [Display omitted] • Evaluating recovery using the Hamilton Depression Rating Scale is difficult. • Biomarkers that indicate a recovery tendency can be used to monitor the effectiveness of drug treatment in MDD. • Several biological processes are associated with drug treatments for depression. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
44. Malaria sporozoites evade host complement attack.
- Author
-
Jiao, Shiming, Tan, Nie, Zhu, Chengyu, Ding, Yan, and Xu, Wenyue
- Subjects
- *
MALARIA , *SPOROZOITES , *COMPLEMENT factor H , *BACTERIAL diseases , *VIRUS diseases , *INTRAVENOUS injections - Abstract
Complement is the first line of the host innate immune response against bacterial and viral infections; however, its role in the development of the malaria liver stage remains undefined. We found that sporozoite infection by either a mosquito bite or intravenous injection activated systemic complement, but neither depletion of C3 nor knockout of C3 had a significant effect on malaria liver stage development. Incubation of mouse serum with trypsin‐treated sporozoites, but not naive sporozoites, led to the deposition of a membrane attack complex (MAC) on the surface of sporozoites and greatly reduced the number of exo‐erythrocytic forms (EEF). Further studies have shown that the recruitment of complement H factor (CFH) may be associated with the prevention of MAC deposition on the surface of naïve sporozoites. Our data strongly suggest that sporozoites can escape complement attacks and provide us with a novel strategy to prevent malaria infection. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
45. A supernumerary synthetic chromosome in Komagataella phaffii as a repository for extraneous genetic material.
- Author
-
Abramczyk, Dariusz, del Carmen Sanchez Olmos, Maria, Rojas, Adan Andres Ramirez, Schindler, Daniel, Robertson, Daniel, McColm, Stephen, Marston, Adele L., and Barlow, Paul N.
- Subjects
- *
ARTIFICIAL chromosomes , *COMPLEMENT factor H , *PROTEIN disulfide isomerase , *HOMOLOGOUS recombination , *HUMAN chromosomes , *GENETIC engineering , *CHROMOSOMES - Abstract
Background: Komagataella phaffii (Pichia pastoris) is a methylotrophic commercially important non-conventional species of yeast that grows in a fermentor to exceptionally high densities on simple media and secretes recombinant proteins efficiently. Genetic engineering strategies are being explored in this organism to facilitate cost-effective biomanufacturing. Small, stable artificial chromosomes in K. phaffii could offer unique advantages by accommodating multiple integrations of extraneous genes and their promoters without accumulating perturbations of native chromosomes or exhausting the availability of selection markers. Results: Here, we describe a linear "nano"chromosome (of 15–25 kb) that, according to whole-genome sequencing, persists in K. phaffii over many generations with a copy number per cell of one, provided non-homologous end joining is compromised (by KU70-knockout). The nanochromosome includes a copy of the centromere from K. phaffii chromosome 3, a K. phaffii-derived autonomously replicating sequence on either side of the centromere, and a pair of K. phaffii-like telomeres. It contains, within its q arm, a landing zone in which genes of interest alternate with long (approx. 1-kb) non-coding DNA chosen to facilitate homologous recombination and serve as spacers. The landing zone can be extended along the nanochromosome, in an inch-worming mode of sequential gene integrations, accompanied by recycling of just two antibiotic-resistance markers. The nanochromosome was used to express PDI, a gene encoding protein disulfide isomerase. Co-expression with PDI allowed the production, from a genomically integrated gene, of secreted murine complement factor H, a plasma protein containing 40 disulfide bonds. As further proof-of-principle, we co-expressed, from a nanochromosome, both PDI and a gene for GFP-tagged human complement factor H under the control of PAOX1 and demonstrated that the secreted protein was active as a regulator of the complement system. Conclusions: We have added K. phaffii to the list of organisms that can produce human proteins from genes carried on a stable, linear, artificial chromosome. We envisage using nanochromosomes as repositories for numerous extraneous genes, allowing intensive engineering of K. phaffii without compromising its genome or weakening the resulting strain. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
46. Complement factor H attenuates TNF-α-induced inflammation by upregulating EIF3C in rheumatoid arthritis.
- Author
-
Jia, Yimeng, Feng, Bin, Ji, Xin, Tian, Xinping, Zhao, Lidan, Zhou, Jiaxin, Zhang, Wen, Li, Mengtao, Fei, Yunyun, and Wu, Xunyao
- Subjects
- *
COMPLEMENT factor H , *C-reactive protein , *RHEUMATOID arthritis , *INFLAMMATORY mediators , *EXPERIMENTAL arthritis , *SYNOVIAL fluid - Abstract
Objective: To explore the role and underlying mechanism of Complement Factor H (CFH) in the peripheral and joint inflammation of RA patients. Methods: The levels of CFH in the serum and synovial fluid were determined by ELISA. The pyroptosis of monocytes was determined by western blotting and flow cytometry. The inflammation cytokine release was tested by ELISA. The cell migration and invasion ability of fibroblast-like synoviocytes (FLS) were tested by Wound healing Assay and transwell assay, respectively. The potential target of CFH was identified by RNA sequencing. Results: CFH levels were significantly elevated in the serum and synovial fluid from RA and associated with high sensitivity C-reactive protein (hs-CRP), erythrocyte sedimentation rate (ESR), and disease activity score 28 (DAS28). TNF-α could inhibit CFH expression, and CFH combined with TNF-α significantly decreased cell death, cleaved-caspase 3, gasdermin E N-terminal (GSDME-N), and inflammatory cytokines release (IL-1β and IL-6) of RA-derived monocytes. Stimulated with TNF-α increased CFH levels in RA FLS and CFH inhibits the migration, invasion, and TNF-α–induced production of inflammatory mediators, including proinflammatory cytokines (IL-6, IL-8) as well as matrix metalloproteinases (MMPs, MMP1 and MMP3) of RA FLSs. The RNA-seq results showed that CFH treatment induced upregulation of eukaryotic translation initiation factor 3 (EIF3C) in both RA monocytes and FLS. The migration of RA FLSs was promoted and the expressions of IL-6, IL-8, and MMP-3 were enhanced upon EIF3C knockdown under the stimulation of CFH combined with TNF-α. Conclusion: In conclusion, we have unfolded the anti-inflammatory roles of CFH in the peripheral and joints of RA, which might provide a potential therapeutic target for RA patients. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
47. Modeling complement activation on human glomerular microvascular endothelial cells.
- Author
-
Stevens, Kes H., Baas, Laura M., van der Velden, Thea J. A. M., Bouwmeester, Romy N., van Dillen, Niels, Dorresteijn, Eiske M., van Zuilen, Arjan D., Wetzels, Jack F. M., Michels, Marloes A. H. M., van de Kar, Nicole C. A. J., and van den Heuvel, Lambertus P.
- Subjects
ECULIZUMAB ,ENDOTHELIAL cells ,COMPLEMENT activation ,COMPLEMENT factor H ,HEMOLYTIC-uremic syndrome ,CHRONIC kidney failure ,CALCIPHYLAXIS ,KIDNEY failure - Abstract
Introduction: Atypical hemolytic uremic syndrome (aHUS) is a rare kidney disease caused by dysregulation of the complement alternative pathway. The complement dysregulation specifically leads to damage to the glomerular endothelium. To further understand aHUS pathophysiology, we validated an ex vivo model for measuring complement deposition on both control and patient human glomerular microvascular endothelial cells (GMVECs). Methods: Endothelial cells were incubated with human test sera and stained with an anti-C5b-9 antibody to visualize and quantify complement depositions on the cells with immunofluorescence microscopy. Results: First, we showed that zymosan-activated sera resulted in increased endothelial C5b-9 depositions compared to normal human serum (NHS). The levels of C5b-9 depositions were similar between conditionally immortalized (ci) GMVECs and primary control GMVECs. The protocol with ciGMVECs was further validated and we additionally generated ciGMVECs from an aHUS patient. The increased C5b-9 deposition on control ciGMVECs by zymosan-activated serum could be dose-dependently inhibited by adding the C5 inhibitor eculizumab. Next, sera from five aHUS patients were tested on control ciGMVECs. Sera from acute disease phases of all patients showed increased endothelial C5b-9 deposition levels compared to NHS. The remission samples showed normalized C5b-9 depositions, whether remission was reached with or without complement blockage by eculizumab. We also monitored the glomerular endothelial complement deposition of an aHUS patient with a hybrid complement factor H (CFH)/CFH-related 1 gene during follow-up. This patient had already chronic kidney failure and an ongoing deterioration of kidney function despite absence of markers indicating an aHUS flare. Increased C5b-9 depositions on ciGMVECs were observed in all samples obtained throughout different diseases phases, except for the samples with eculizumab levels above target. We then tested the samples on the patient's own ciGMVECs. The C5b-9 deposition pattern was comparable and these aHUS patient ciGMVECs also responded similar to NHS as control ciGMVECs. Discussion: In conclusion, we demonstrate a robust and reliable model to adequately measure C5b-9-based complement deposition on human control and patient ciGMVECs. This model can be used to study the pathophysiological mechanisms of aHUS or other diseases associated with endothelial complement activation ex vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
48. Identification of complement factor H variants that predispose to pre‐eclampsia: A genetic and functional study.
- Author
-
Lokki, A. Inkeri, Ren, Zhen, Triebwasser, Michael, Daly, Emma, Perola, Markus, Auro, Kirsi, Burwick, Richard, Salmon, Jane E., Daly, Mark, Laivuori, Hannele, Atkinson, John P., Java, Anuja, Meri, Seppo, Heinonen, Seppo, Kajantie, Eero, Kere, Juha, Kivinen, Katja, and Pouta, Anneli
- Subjects
- *
COMPLEMENT factor H , *ECLAMPSIA , *PREECLAMPSIA , *COMPLEMENT activation , *GENETIC variation , *MISSENSE mutation - Abstract
Objective: The objective of the study was to investigate the role of genetic variants in complement proteins in pre‐eclampsia. Design: In a case–control study involving 609 cases and 2092 controls, five rare variants in complement factor H (CFH) were identified in women with severe and complicated pre‐eclampsia. No variants were identified in controls. Setting: Pre‐eclampsia is a leading cause of maternal and fetal morbidity and mortality. Immune maladaptation, in particular, complement activation that disrupts maternal‐fetal tolerance leading to placental dysfunction and endothelial injury, has been proposed as a pathogenetic mechanism, but this remains unproven. Population: We genotyped 609 pre‐eclampsia cases and 2092 controls from FINNPEC and the national FINRISK cohorts. Methods: Complement‐based functional and structural assays were conducted in vitro to define the significance of these five missense variants and each compared with wild type. Main outcome measures: Secretion, expression and ability to regulate complement activation were assessed for factor H proteins harbouring the mutations. Results: We identified five heterozygous rare variants in complement factor H (L3V, R127H, R166Q, C1077S and N1176K) in seven women with severe pre‐eclampsia. These variants were not identified in controls. Variants C1077S and N1176K were novel. Antigenic, functional and structural analyses established that four (R127H, R166Q, C1077S and N1176K) were deleterious. Variants R127H and C1077S were synthesised, but not secreted. Variants R166Q and N1176K were secreted normally but showed reduced binding to C3b and consequently defective complement regulatory activity. No defect was identified for L3V. Conclusions: These results suggest that complement dysregulation due to mutations in complement factor H is among the pathophysiological mechanisms underlying severe pre‐eclampsia. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
49. C-reactive protein-complement factor H axis as a biomarker of activity in early and intermediate age-related macular degeneration
- Author
-
Lena Giralt, Marc Figueras-Roca, Beatriz De Luis Eguileor, Barbara Romero, Javier Zarranz-Ventura, Socorro Alforja, Francisca Santiago, Jennifer Bolaños, Francisco Lozano, Marina Dotti-Boada, Anna Sala-Puigdollers, Paula Dura, Jordi Izquierdo-Serra, Oliver Valero, Alfredo Adan, Alex Fonollosa, and Blanca Molins
- Subjects
age-related macular degeneration ,inflammation ,complement factor H ,C-reactive protein ,retina ,biomarker ,Immunologic diseases. Allergy ,RC581-607 - Abstract
PurposeTo determine and compare the serum levels of complement Factor H (FH), monomeric C-Reactive Protein (mCRP) and pentameric C-Reactive protein (pCRP) in patients with age-related macular degeneration (AMD) and to correlate them with clinical, structural and functional parameters.MethodsCross-sectional observational study. One hundred thirty-nine individuals (88 patients and 51 healthy controls) from two referral centers were included and classified into three groups: early or intermediate AMD (n=33), advanced AMD (n=55), and age and sex matched healthy controls (n=51). Serum levels of FH, mCRP, and pCRP were determined and correlated with clinical and imaging parameters.ResultsPatients with intermediate AMD presented FH levels significantly lower than controls [186.5 (72.1-931.8) µg/mL vs 415.2 (106.1-1962.2) µg/mL; p=0.039] and FH levels
- Published
- 2024
- Full Text
- View/download PDF
50. Complement factor H: a novel innate immune checkpoint in cancer immunotherapy
- Author
-
Ruchi Saxena, Elizabeth B. Gottlin, Michael J. Campa, Ryan T. Bushey, Jian Guo, Edward F. Patz, and You-Wen He
- Subjects
complement ,complement factor H ,immunosuppression ,tumor micro-environment ,immune checkpoint ,immunotherapy ,Biology (General) ,QH301-705.5 - Abstract
The elimination of cancer cells critically depends on the immune system. However, cancers have evolved a variety of defense mechanisms to evade immune monitoring, leading to tumor progression. Complement factor H (CFH), predominately known for its function in inhibiting the alternative pathway of the complement system, has recently been identified as an important innate immunological checkpoint in cancer. CFH-mediated immunosuppression enhances tumor cells’ ability to avoid immune recognition and produce an immunosuppressive tumor microenvironment. This review explores the molecular underpinnings, interactions with immune cells, clinical consequences, and therapeutic possibilities of CFH as an innate immune checkpoint in cancer control. The difficulties and opportunities of using CFH as a target in cancer immunotherapy are also explored.
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.