Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Mol Immunol ; 168: 10-16, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38368725

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.


Subject(s)
Complement Factor H/deficiency , Glomerulonephritis, Membranoproliferative , Hereditary Complement Deficiency Diseases , Kidney Diseases , Humans , Animals , Mice , Complement C3/genetics , Complement C3/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Complement Factor H/genetics , Complement Factor H/therapeutic use , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/drug therapy , Glomerulonephritis, Membranoproliferative/metabolism , Complement Pathway, Alternative
2.
J Mol Med (Berl) ; 102(4): 571-583, 2024 04.
Article in English | MEDLINE | ID: mdl-38418621

ABSTRACT

Ankylosing spondylitis (AS) is a chronic inflammatory disease, characterized by excessive new bone formation. We previously reported that the complement factor H-related protein-5 (CFHR5), a member of the human factor H protein family, is significantly elevated in patients with AS compared to other rheumatic diseases. However, the pathophysiological mechanism underlying new bone formation by CFHR5 is not fully understood. In this study, we revealed that CFHR5 and proinflammatory cytokines (TNF, IL-6, IL-17A, and IL-23) were elevated in the AS group compared to the HC group. Correlation analysis revealed that CFHR5 levels were not significantly associated with proinflammatory cytokines, while CFHR5 levels in AS were only positively correlated with the high CRP group. Notably, treatment with soluble CFHR5 has no effect on clinical arthritis scores and thickness at hind paw in curdlan-injected SKG, but significantly increased the ectopic bone formation at the calcaneus and tibia bones of the ankle as revealed by micro-CT image and quantification. Basal CFHR5 expression was upregulated in AS-osteoprogenitors compared to control cells. Also, treatment with CFHR5 remarkedly induced bone mineralization status of AS-osteoprogenitors during osteogenic differentiation accompanied by MMP13 expression. We provide the first evidence demonstrating that CFHR5 can exacerbate the pathological bone formation of AS. Therapeutic modulation of CFHR5 could be promising for future treatment of AS. KEY MESSAGES: Serum level of CFHR5 is elevated and positively correlated with high CRP group of AS patients. Recombinant CFHR5 protein contributes to pathological bone formation in in vivo model of AS. CFHR5 is highly expressed in AS-osteoprogenitors compared to disease control. Recombinant CFHR5 protein increased bone mineralization accompanied by MMP13 in vitro model of AS.


Subject(s)
Spondylitis, Ankylosing , Humans , Complement Factor H/therapeutic use , Complement System Proteins/metabolism , Cytokines , Matrix Metalloproteinase 13 , Osteogenesis , Spondylitis, Ankylosing/pathology
3.
J Transl Med ; 21(1): 846, 2023 11 23.
Article in English | MEDLINE | ID: mdl-37996918

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-1ß 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.


Subject(s)
Arthritis, Rheumatoid , Tumor Necrosis Factor-alpha , Humans , Arthritis, Rheumatoid/drug therapy , Cell Proliferation , Cells, Cultured , Complement Factor H/genetics , Complement Factor H/metabolism , Complement Factor H/therapeutic use , Cytokines/metabolism , Fibroblasts/metabolism , Inflammation/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Synovial Membrane/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Mol Cancer Ther ; 22(6): 778-789, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36995981

ABSTRACT

Development of novel therapeutic antibodies that not only kill tumor cells but modulate the adaptive immune response has the potential to produce long term anticancer immunity and a durable clinical response. We previously reported the discovery of anti-complement factor H (CFH) autoantibodies in patients with lung cancer that were associated with early-stage disease and exceptional outcomes. The human mAb GT103, produced from a single CFH autoantibody-expressing B cell of a patient with lung cancer, recognizes a conformationally distinct epitope on tumor cells, kills tumor cells, and inhibits tumor growth in animal studies. Recent experiments have shown that GT103 restructures the tumor microenvironment and initiates a robust antitumoral adaptive immune response. The current study further elucidates several mechanisms by which GT103 kills tumor cells and drives the immune program. Here we show GT103 has specificity for tumor cells without binding to native soluble CFH or normal tissues. GT103 causes complement C3 split product deposition on tumor cells in vitro and in vivo, triggers antibody-dependent cellular phagocytosis, and increases translocation of the danger-associated molecular pattern molecule calreticulin to the plasma membrane. We also demonstrate that GT103 causes B-cell activation in vitro and in vivo, and that GT103 antitumor activity in vivo is B-cell dependent. The complex mechanism of GT103, a tumor-specific antibody that kills tumor cells and stimulates an immune response, supports further development of this human-derived antibody as a novel therapeutic option for patients with lung cancer.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Animals , Humans , Complement Factor H/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Lung Neoplasms/pathology , Autoantibodies/therapeutic use , Antineoplastic Agents/therapeutic use , Tumor Microenvironment
5.
Int J Hematol ; 116(5): 812-818, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35790695

ABSTRACT

Complement-mediated hemolytic uremic syndrome (CM-HUS) following chemotherapy for pediatric acute lymphoid neoplasms has rarely been reported. We report the case of an 8-year-old boy with T-lymphoblastic lymphoma (T-LBL) who developed CM-HUS with complement factor H (CFH) mutations (S1191L, V1197A) during induction therapy. Safe administration of chemotherapy after CM-HUS recovery was challenging. By closely monitoring hemolytic and renal parameters during the 2-year treatment period, we observed four episodes of microangiopathic hemolytic anemia (MAHA) with hypocomplementemia and low haptoglobin but no renal dysfunction or thrombocytopenia. Here, we describe the MAHA and CM-HUS episodes in the hopes of elucidating the complex pathophysiology of disorders associated with CFH mutation.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Hemolytic-Uremic Syndrome , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Purpura, Thrombotic Thrombocytopenic , Male , Humans , Child , Complement Factor H/genetics , Complement Factor H/therapeutic use , Hemolysis , Haptoglobins/therapeutic use , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/pathology , Hemolytic-Uremic Syndrome/therapy , Purpura, Thrombotic Thrombocytopenic/therapy , Complement System Proteins , Atypical Hemolytic Uremic Syndrome/drug therapy , Atypical Hemolytic Uremic Syndrome/genetics
6.
Ren Fail ; 44(1): 1061-1069, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35730179

ABSTRACT

OBJECTIVE: To explore the clinical characteristics, treatment protocol and prognosis of children with anti-complement factor H (CFH) autoantibody (Ab)-associated hemolytic uremic syndrome (HUS). METHODS: Clinical data of 8 patients with anti-CFH Ab-associated HUS who were admitted to Shandong Provincial Hospital from January 2011 to December 2020 were collected retrospectively. RESULTS: The age at disease onset ranged between 5.83 and 13.5 years, with a male: female ratio of 1.67:1. The time of onset was distributed from May to June and November to December. Digestive and upper respiratory tract infections were common prodromal infections. Positivity for anti-CFH Ab and reduced C3 levels were observed among all patients. Heterozygous mutation of the CHFR5 gene (c.669del A) and homozygous loss of the CFHR1 gene [loss2(EXON:2-6)] were found in two patients. All patients received early treatment with plasma exchange and corticosteroid therapy. Six patients were given immunosuppressive agents (cyclophosphamide and/or mycophenolate mofetil) for persistent proteinuria. The follow-up period was 12-114 months. Four of 8 patients achieved complete remission, 3 achieved partial remission, and 1 died. Relapse occurred in two patients. CONCLUSION: Children with anti-CFH Ab-associated HUS were mainly school-aged and predominantly male, with onset times of summer and winter. Digestive and upper respiratory tract infections were common prodromal infections. Plasma exchange combined with methylprednisolone pulse therapy in the acute phase and cyclophosphamide or mycophenolate mofetil treatment for maintenance can be utilized in children with anti-CFH Ab-associated HUS if eculizumab is not available.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Hemolytic-Uremic Syndrome , Respiratory Tract Infections , Adolescent , Atypical Hemolytic Uremic Syndrome/drug therapy , Atypical Hemolytic Uremic Syndrome/therapy , Autoantibodies , Child , Child, Preschool , Complement Factor H/genetics , Complement Factor H/therapeutic use , Cyclophosphamide/therapeutic use , Female , Hemolytic-Uremic Syndrome/complications , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/therapy , Humans , Immunologic Factors/therapeutic use , Male , Mycophenolic Acid/therapeutic use , Respiratory Tract Infections/complications , Respiratory Tract Infections/drug therapy , Retrospective Studies
7.
Adv Mater ; 34(8): e2107070, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34910334

ABSTRACT

Complement opsonization is among the biggest challenges facing nanomedicine. Nearly instantly after injection into blood, nanoparticles are opsonized by the complement protein C3, leading to clearance by phagocytes, fouling of targeting moieties, and release of anaphylatoxins. While surface polymers such as poly(ethylene glycol) (PEG) partially decrease complement opsonization, most nanoparticles still suffer from extensive complement opsonization, especially when linked to targeting moieties. To ameliorate the deleterious effects of complement, two of mammals' natural regulators of complement activation (RCAs), Factors H and I, are here conjugated to the surface of nanoparticles. In vitro, Factor H or I conjugation to PEG-coated nanoparticles decrease their C3 opsonization, and markedly reduce nanoparticle uptake by phagocytes. In an in vivo mouse model of sepsis-induced lung injury, Factor I conjugation abrogates nanoparticle uptake by intravascular phagocytes in the lungs, allowing the blood concentration of the nanoparticle to remain elevated much longer. For nanoparticles targeted to the lung's endothelium by conjugation to anti-ICAM antibodies, Factor I conjugation shifts the cell-type distribution away from phagocytes and toward endothelial cells. Finally, Factor I conjugation abrogates the severe anaphylactoid responses common to many nanoparticles, preventing systemic capillary leak and preserving blood flow to visceral organs and the brain. Thus, conjugation of RCAs, like Factor I, to nanoparticles is likely to help in nanomedicine's long battle against complement, improving several key parameters critical for clinical success.


Subject(s)
Complement C3 , Nanomedicine , Nanoparticles , Animals , Complement Activation , Complement C3/metabolism , Complement C3/pharmacology , Complement Factor H/therapeutic use , Endothelial Cells/metabolism , Fibrinogen/therapeutic use , Mammals/metabolism , Mice , Nanomedicine/methods , Nanoparticles/adverse effects , Nanoparticles/therapeutic use , Opsonization
8.
J Immunol ; 199(11): 3828-3839, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29084837

ABSTRACT

Bacteria can cause life-threatening infections, such as pneumonia, meningitis, or sepsis. Antibiotic therapy is a mainstay of treatment, although antimicrobial resistance has drastically increased over the years. Unfortunately, safe and effective vaccines against most pathogens have not yet been approved, and thus developing alternative treatments is important. We analyzed the efficiency of factor H (FH)6-7/Fc, a novel antibacterial immunotherapeutic protein against the Gram-positive bacterium Streptococcus pyogenes This protein is composed of two domains of complement inhibitor human FH (FH complement control protein modules 6 and 7) that bind to S. pyogenes, linked to the Fc region of IgG (FH6-7/Fc). FH6-7/Fc has previously been shown to enhance complement-dependent killing of, and facilitate bacterial clearance in, animal models of the Gram-negative pathogens Haemophilus influenzae and Neisseria meningitidis We hypothesized that activation of complement by FH6-7/Fc on the surface of Gram-positive bacteria such as S. pyogenes will enable professional phagocytes to eliminate the pathogen. We found that FH6-7/Fc alleviated S. pyogenes-induced sepsis in a transgenic mouse model expressing human FH (S. pyogenes binds FH in a human-specific manner). Furthermore, FH6-7/Fc, which binds to protein H and selected M proteins, displaced FH from the bacterial surface, enhanced alternative pathway activation, and reduced bacterial blood burden by opsonophagocytosis in a C3-dependent manner in an ex vivo human whole-blood model. In conclusion, FH-Fc chimeric proteins could serve as adjunctive treatments against multidrug-resistant bacterial infections.


Subject(s)
Complement Factor H/therapeutic use , Immunotherapy/methods , Recombinant Fusion Proteins/therapeutic use , Sepsis/therapy , Staphylococcal Vaccines/immunology , Streptococcal Infections/therapy , Streptococcus pyogenes/immunology , Animals , Anti-Bacterial Agents/therapeutic use , Cells, Cultured , Complement C3/metabolism , Complement C3 Convertase, Alternative Pathway , Complement Factor H/genetics , Drug Resistance, Multiple , Humans , Mice , Mice, Transgenic , Phagocytosis , Recombinant Fusion Proteins/genetics , Sepsis/immunology , Streptococcal Infections/immunology
9.
J Am Soc Nephrol ; 28(5): 1462-1474, 2017 May.
Article in English | MEDLINE | ID: mdl-27932477

ABSTRACT

Genetic defects in complement regulatory proteins can lead to severe renal diseases, including atypical hemolytic uremic syndrome and C3 glomerulopathies, and age-related macular degeneration. The majority of the mutations found in patients with these diseases affect the glycoprotein complement factor H, the main regulator of the alternative pathway of complement activation. Therapeutic options are limited, and novel treatments, specifically those targeting alternative pathway activation, are highly desirable. Substitution with biologically active factor H could potentially treat a variety of diseases that involve increased alternative pathway activation, but no therapeutic factor H is commercially available. We recently reported the expression of full-length recombinant factor H in moss (Physcomitrella patens). Here, we present the production of an improved moss-derived recombinant human factor H devoid of potentially immunogenic plant-specific sugar residues on protein N-glycans, yielding approximately 1 mg purified moss-derived human factor H per liter of initial P. patens culture after a multistep purification process. This glycosylation-optimized factor H showed full in vitro complement regulatory activity similar to that of plasma-derived factor H and efficiently blocked LPS-induced alternative pathway activation and hemolysis induced by sera from patients with atypical hemolytic uremic syndrome. Furthermore, injection of moss-derived factor H reduced C3 deposition and increased serum C3 levels in a murine model of C3 glomerulopathy. Thus, we consider moss-produced recombinant human factor H a promising pharmaceutical product for therapeutic intervention in patients suffering from complement dysregulation.


Subject(s)
Atypical Hemolytic Uremic Syndrome/drug therapy , Bryopsida , Complement System Proteins , Immune System Diseases/drug therapy , Animals , Bryopsida/metabolism , Complement Factor H/biosynthesis , Complement Factor H/metabolism , Complement Factor H/therapeutic use , Glycosylation , Humans , Mice
10.
PLoS One ; 11(2): e0149307, 2016.
Article in English | MEDLINE | ID: mdl-26872035

ABSTRACT

Streptococcus pneumoniae is a common cause of sepsis. Effective complement activation is an important component of host defence against invading pathogens, whilst excessive complement activation has been associated with endothelial dysfunction and organ damage. The alternative pathway amplification loop is important for the enhancement of complement activation. Factor H is a key negative regulator of the alternative pathway amplification loop and contributes to tight control of complement activation. We assessed the effect of inhibition of the alternative pathway on sepsis associated inflammation and disease severity using human factor H treatment in a clinically relevant mice model of pneumococcal sepsis. Mice were infected intravenously with live Streptococcus pneumoniae. At the first clinical signs of infection, 17 hours post-infection, mice were treated with ceftriaxone antibiotic. At the same time purified human factor H or in controls PBS was administered. Treatment with human factor H did not attenuate disease scores, serum pro-inflammatory cytokines, or vascular permeability and did not significantly affect C3 and C3a production at 26 h post-infection. Therefore, we conclude that inhibition of the alternative complement pathway by exogenous human factor H fails to attenuate inflammation and vascular leakage at a clinically relevant intervention time point in pneumococcal sepsis in mice.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Capillary Permeability/drug effects , Ceftriaxone/therapeutic use , Pneumococcal Infections/drug therapy , Sepsis/drug therapy , Streptococcus pneumoniae/drug effects , Animals , Complement Factor H/therapeutic use , Cytokines/blood , Cytokines/immunology , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Pneumococcal Infections/blood , Pneumococcal Infections/complications , Pneumococcal Infections/immunology , Sepsis/blood , Sepsis/complications , Sepsis/immunology , Streptococcus pneumoniae/immunology
11.
J Gene Med ; 17(10-12): 229-43, 2015.
Article in English | MEDLINE | ID: mdl-26369397

ABSTRACT

BACKGROUND: Age-related macular degeneration (AMD) is the most common cause of blindness in the elderly, with no therapy available for 90% of patients. Recent genetic evidence implicates activation of complement in the pathogenesis of AMD. We have recently discovered that adenovirus (Ad)-mediated expression of complement component C3 (AdCMVC3) in the murine retina recapitulates many of the pathological features found in human AMD. In the present study, utilizing a gene therapy approach, we examine whether Ad-mediated expression of complement Factor H (AdCAGfH) attenuates AdCMVC3-mediated retinal pathology. METHODS: AdCMVC3 was co-injected with either AdCAGfH or a negative control virus expressing green fluorescent protein (AdCMVGFP) into the subretinal space of adult mice. The resulting retinal pathology was analyzed by histology and immunocytochemistry and retinal function was quantified by electroretinography. RESULTS: Morphological and functional analyses indicated that AdCMVC3-mediated retinal pathology could be attenuated by AdCAGfH. Specifically, endothelial cell proliferation was reduced by 91% and atrophy of retinal pigment epithelium (RPE) could be attenuated by 69%. AdCAGfH injected eyes exhibited 90-150% greater A-wave and 120-180% greater B-wave amplitudes relative to control eyes. Immunocytochemical analysis of rhodopsin and RPE65 was consistent with the rescue of photoreceptors and RPE in AdCAGfH injected eyes. CONCLUSIONS: C3-induced pathology in murine retina can be attenuated by Ad-mediated expression of Factor H. Expression of Factor H is worthy of further study as a potential gene therapy for AMD.


Subject(s)
Complement C3/metabolism , Complement Factor H/therapeutic use , Adenoviridae , Animals , Complement C3/adverse effects , Complement Factor H/administration & dosage , Genetic Therapy , Genetic Vectors , Humans , Macular Degeneration/therapy , Mice , Retina/drug effects , Retina/pathology
12.
Vestn Oftalmol ; 129(5): 128-35, 2013.
Article in Russian | MEDLINE | ID: mdl-24261289

ABSTRACT

A study on the role of CFH, HTRA and IL-8 gene polymorphism in age-related macular degeneration (AMD) development has been conducted. At the first stage of the study genetic testing was done in 69 patients with exudative AMD and 370 random Moscow citizens without the disease. The goal of the second stage was to determine the influence of gene polymorphism on patient's response to endovitreal ranibizumab treatment. For that, visual acuity and foveal thickness were assessed before and after ranibizumab injections in 120 patients with wet AMD. All patients were genotyped for the genes of interest. The results showed that the presence of homozygous 402H polymorphism in CFH gene, as well as homozygous (-625)A mutation in HTRA1 gene, determines certain clinical presentations. Moreover, visual acuity below 0.1 and presence of 402H, (-625)A and (-251)A alleles in both copies of all three genes (CFH, HTRA and IL-8) are negative predictors of disease severity and antiangiogenic treatment response.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Complement Factor H/therapeutic use , Interleukin-8/therapeutic use , Macular Degeneration/genetics , Pharmacogenetics/methods , Polymorphism, Genetic , Serine Endopeptidases/therapeutic use , Alleles , Complement Factor H/genetics , Complement Inactivating Agents/therapeutic use , DNA/genetics , Female , Fluorescein Angiography , Fundus Oculi , High-Temperature Requirement A Serine Peptidase 1 , Homozygote , Humans , Interleukin-8/genetics , Intravitreal Injections , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Male , Ranibizumab , Serine Endopeptidases/genetics , Visual Acuity
13.
Arch. Soc. Esp. Oftalmol ; 88(1): 3-10, ene. 2013. tab
Article in Spanish | IBECS | ID: ibc-109503

ABSTRACT

Propósito: Demostrar la influencia genética en el desarrollo de degeneración macular asociada a la edad (DMAE) analizando las distribuciones genotípicas de haplotipos de polimorfismos de genes con relación demostrada con la aparición de DMAE (CFH, ARMS2, HTRA1) en pacientes con DMAE y personas sanas. Método: Se tomaron 101 pacientes diagnosticados de DMAE (74 exudativa y 27 atrófica) según las normas del sistema internacional de clasificación Wisconsin. Como control se tomaron 91 pacientes sin DMAE ni otras alteraciones maculares. Se analizó el polimorfismo rs 1410996 del gen CFH, el rs 10940923 de ARMS2 mediante PCR a tiempo real con sondas Taqman y el HTRA1 -625 mediante digestión con endonucleasas de restricción .Se estudió la presencia de haplotipos que combinaban los genotipos que habían demostrado aumentar el riesgo de DMAE de los polimorfismos estudiados de CFH, HTRA1 y ARMS2 en estudios previos en nuestro grupo de pacientes y el grupo control. Resultados: Se demostró que es más frecuente en el grupo de pacientes, de forma estadísticamente significativa, la expresión simultánea de los genotipos GG de CFH (rs 1410996) y TT de ARMS2 (rs 10940923) (p=0,037; OR: 7,742 [1,010-63,156]); TT de ARMS2 (rs 10940923) y GG de HTRA1-625 (p=0,001; OR: 9,006 [2,019-40,168]) y GG de CFH (rs1410996), TT de ARMS2 (rs 1040923) y GG de HTRA1 -625 (p=0,043; OR: 6,702 [1,003-55,565]). Conclusiones: La presencia de haplotipos que combinan genotipos, considerados de riesgo en estudios previos, de los polimorfismos analizados es más frecuente en pacientes con DMAE y parece aumentar el riesgo de padecer la enfermedad en nuestra población(AU)


Objective: To demonstrate genetic influence on the onset of age-related macular disease (AMD), analyzing genotype distribution of haplotypes, including polymorphisms of genes with proved relationships with AMD risk (CFH, ARMS2, HTRA1) in patients with AMD and in healthy people. Methods: We took 101 consecutive patients with an AMD diagnosis following Wisconsin international classification. For our control group, we took 91 patients without AMD or any significant macular changes. We analyzed CFH rs 1410996, ARMS2 rs 10940923 polymorphisms using real time PCR with taqman probes, and HTRA1 -625 using restriction endonuclease digestion. We studied haplotypes by simultaneously combining genotypes which, in previous studies, had been shown to have relationship with AMD (CFH, ARMS2, HTRA1) in patients with AMD and healthy people. Results: There was a statistically significant higher proportion of patients with AMD simultaneously expressing CFH GG (rs 1410996) and ARMS2 TT (rs 10940923) (P=0.037; OR: 7.742 [1.010-63.156]); ARMS2 TT (rs 10940923) and HTRA1-625 TT (P=0.001; OR: 9.006 [2.019-40.168]) and CFH GG (rs 1410996), ARMS2 TT (rs 1040923) and HTRA1 -625 GG (P=0.043; OR: 6.702 [1.003-55.565]) genotypes. Conclusions: Haplotypes which combine «risk genotypes», demonstrated in previous studies, of our anlyzed polymorphisms are more frequent in patients with AMD than in the control group, and they seem to increase the risk of suffering the disease in our population(AU)


Subject(s)
Humans , Male , Female , Macular Degeneration/complications , Macular Degeneration/diagnosis , Haplotypes , Haplotypes/physiology , Genotyping Techniques/instrumentation , Genotyping Techniques/methods , Genotyping Techniques , Complement Factor H , Complement Factor H/therapeutic use , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Macular Degeneration/physiopathology , Macular Degeneration , Genotyping Techniques/trends
14.
Biol Chem ; 393(6): 429-39, 2012 May.
Article in English | MEDLINE | ID: mdl-22628306

ABSTRACT

Chronic neurodegenerative disorders and acute injuries of the central nervous system exert a prohibitive economic burden, which is aggravated by an unmet medical need for the development of effective neurotherapeutics. The evolutionarily conserved neuropeptide, adrenomedullin (AM), and its binding protein, AMBP-1, also known as complement factor H, play important roles in brain physiology, and their expression is altered in brain pathology. In this review, we discuss the molecular regulation of AM and AMBP-1 and the pivotal roles they play in neuroprotection following brain injury. We assess the reciprocal synergistic effects of AM and AMBP-1 and make suggestions for the design of a novel combination neurotherapy devoid of the potential hypotensive effects of AM while optimizing its neuroprotective property.


Subject(s)
Adrenomedullin/metabolism , Brain/metabolism , Complement Factor H/metabolism , Neuroprotective Agents/metabolism , Adrenomedullin/genetics , Adrenomedullin/pharmacology , Adrenomedullin/therapeutic use , Animals , Brain/drug effects , Brain Injuries/drug therapy , Brain Injuries/metabolism , Complement Factor H/genetics , Complement Factor H/pharmacology , Complement Factor H/therapeutic use , Humans , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
15.
J Ocul Pharmacol Ther ; 28(4): 402-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22309197

ABSTRACT

PURPOSE: Genetic associations and the presence of complement components within pathological structures of age-related macular degeneration (AMD) have generated the hypothesis that AMD is caused by chronic local complement activation. Since the majority of activity in the common terminal pathway results from engagement of the amplification loop, the alternative pathway has been proposed as a logical therapeutic target. We recently generated a factor H (fH)-based complement inhibitor (CR2-fH) with the capacity to be "targeted" to sites of complement C3 activation. We asked whether the human therapeutic (TT30) is effective in a mouse model of AMD. METHODS: Choroidal neovascularization (CNV) was induced by argon laser photocoagulation of Bruch's membrane. Every other day, mice received intravenous injections of TT30 or vehicles, and after 6 days, the presence or absence of CNV and CNV-related changes were evaluated. Area of CNV, photoreceptor cell function, gene expression for complement components and cytokines, vascular endothelial growth factor (VEGF) protein levels, and TT30 bioavailability were determined. RESULTS: CNV development, which has previously been shown to require local complement activation, could be reduced by intravenous TT30 delivery. Specific inhibition of the alternative pathway not only reduced angiogenesis in CNV, but also ameliorated changes in several associated disease-related biomarkers, including diminished retinal function and molecular events known to be involved in AMD such as VEGF production. After intravenous injection, TT30 localized to CNV lesion sites in the retinal pigmented epithelium-choroid. CONCLUSION: Systemic administration of TT30 was found to reduce CNV pathology. These data may open new avenues for novel systemic AMD treatment strategies.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Choroidal Neovascularization/prevention & control , Complement Factor H/therapeutic use , Complement Pathway, Alternative/drug effects , Lasers/adverse effects , Receptors, Complement 3d/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Animals , Biomarkers , Choroidal Neovascularization/pathology , Complement Factor H/biosynthesis , Electroretinography , Humans , Immunohistochemistry , Ligands , Mice , Mice, Inbred C57BL , Plasmids , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Complement 3d/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/metabolism
16.
Blood ; 118(17): 4705-13, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21860027

ABSTRACT

To selectively modulate human complement alternative pathway (CAP) activity implicated in a wide range of acute and chronic inflammatory conditions and to provide local cell surface and tissue-based inhibition of complement-induced damage, we developed TT30, a novel therapeutic fusion protein linking the human complement receptor type 2 (CR2/CD21) C3 fragment (C3frag = iC3b, C3dg, C3d)-binding domain with the CAP inhibitory domain of human factor H (fH). TT30 efficiently blocks ex vivo CAP-dependent C3frag accumulation on activated surfaces, membrane attack complex (MAC) formation and hemolysis of RBCs in a CR2-dependent manner, and with a ∼ 150-fold potency gain over fH, without interference of C3 activation or MAC formation through the classic and lectin pathways. TT30 protects RBCs from hemolysis and remains bound and detectable for at least 24 hours. TT30 selectively inhibits CAP in cynomolgus monkeys and is bioavailable after subcutaneous injection. Using a unique combination of targeting and effector domains, TT30 controls cell surface CAP activation and has substantial potential utility for the treatment of human CAP-mediated diseases.


Subject(s)
Complement C3-C5 Convertases/antagonists & inhibitors , Complement C3d/metabolism , Complement Factor H/therapeutic use , Complement Pathway, Alternative/immunology , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/therapeutic use , Immune System Diseases/drug therapy , Immune System Diseases/immunology , Receptors, Complement 3d/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Animals , Complement C3-C5 Convertases/metabolism , Complement Factor H/administration & dosage , Drug Design , Drug Evaluation, Preclinical , Female , Humans , Immune System Diseases/metabolism , Macaca fascicularis , Male , Models, Immunological , Molecular Targeted Therapy/methods , Rabbits , Receptors, Complement 3d/administration & dosage , Recombinant Fusion Proteins/administration & dosage
17.
J Immunol ; 186(7): 4269-77, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21339361

ABSTRACT

Exposure of nonself surfaces such as those of biomaterials or transplanted cells and organs to host blood frequently triggers innate immune responses, thereby affecting both their functionality and tolerability. Activation of the alternative pathway of complement plays a decisive role in this unfavorable reaction. Whereas previous studies demonstrated that immobilization of physiological regulators of complement activation (RCA) can attenuate this foreign body-induced activation, simple and efficient approaches for coating artificial surfaces with intact RCA are still missing. The conjugation of small molecular entities that capture RCA with high affinity is an intriguing alternative, as this creates a surface with autoregulatory activity upon exposure to blood. We therefore screened two variable cysteine-constrained phage-displayed peptide libraries for factor H-binding peptides. We discovered three peptide classes that differed with respect to their main target binding areas. Peptides binding to the broad middle region of factor H (domains 5-18) were of particular interest, as they do not interfere with either regulatory or binding activities. One peptide in this group (5C6) was further characterized and showed high factor H-capturing activity while retaining its functional integrity. Most importantly, when 5C6 was coated to a model polystyrene surface and exposed to human lepirudin-anticoagulated plasma, the bound peptide captured factor H and substantially inhibited complement activation by the alternative pathway. Our study therefore provides a promising and novel approach to produce therapeutic materials with enhanced biocompatibility.


Subject(s)
Complement Pathway, Alternative/immunology , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , Biocompatible Materials/metabolism , Cloning, Molecular , Complement C3b/antagonists & inhibitors , Complement C3b/metabolism , Complement Factor H/metabolism , Complement Factor H/therapeutic use , Complement Factor I/antagonists & inhibitors , Complement Factor I/metabolism , Complement Inactivator Proteins/metabolism , Hemolysis , Humans , Peptide Library , Protein Binding/immunology , Surface Properties
18.
J Immunol ; 185(11): 7007-13, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20962256

ABSTRACT

Ischemia reperfusion injury (IRI) is an unavoidable event during solid organ transplantation and is a major contributor to early graft dysfunction and subsequent graft immunogenicity. In a therapeutic paradigm using targeted complement inhibitors, we investigated the role of complement, and specifically the alternative pathway of complement, in IRI to heart isografts. Mouse heterotopic isograft heart transplants were performed in C57BL/6 mice treated with a single injection of either CR2-Crry (inhibits all complement pathways) or CR2-fH (inhibits alternative complement pathway) immediately posttransplantation. Transplanted hearts were harvested at 12 and 48 h for analysis. Both inhibitors resulted in a significant reduction in myocardial IRI, as measured by histology and serum cardiac troponin I levels. Furthermore, compared with untreated controls, both inhibitors reduced graft complement deposition, neutrophil and macrophage infiltration, adhesion molecule expression (P-selectin, E-selectin, and I-CAM-1), and proinflammatory cytokine expression (TNF-α, IL-1ß, KC, and MCP-1). The reduction in myocardial damage and cellular infiltration was not significantly different between CR2-Crry- and CR2-fH-treated mice, although adhesion molecule and cytokine levels were significantly lower in CR2-Crry-treated mice compared with CR2-fH-treated mice. In conclusion, the alternative complement pathway plays a major contributing role in myocardial IRI after heart transplantation, and local (targeted) complement inhibition has the potential to provide an effective and safe therapeutic strategy to reduce graft injury. Although total complement blockade may be somewhat more efficacious in terms of reducing inflammation, specific blockade of the alternative pathway is likely to be less immunosuppressive in an already immunocompromised recipient.


Subject(s)
Complement Factor H/physiology , Complement Inactivator Proteins/physiology , Complement Pathway, Alternative/immunology , Heart Transplantation/immunology , Myocardial Reperfusion Injury/prevention & control , Recombinant Fusion Proteins/physiology , Animals , Complement Factor H/therapeutic use , Complement Inactivator Proteins/therapeutic use , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Drug Delivery Systems/methods , Heart Transplantation/pathology , Leukocyte Count , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Random Allocation , Receptors, Complement 3d/physiology , Receptors, Complement 3d/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Transplantation, Homologous
19.
J Surg Res ; 163(1): 110-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20538296

ABSTRACT

BACKGROUND: Acute renal failure secondary to ischemia and reperfusion (I/R) injury poses a significant burden on both surgeons and patients. It carries a high morbidity and mortality rate and no specific treatment currently exists. Major causes of renal I/R injury include trauma, sepsis, hypoperfusion, and various surgical procedures. We have demonstrated that adrenomedullin (AM), a novel vasoactive peptide, combined with AM binding protein-1 (AMBP-1), which augments the activity of AM, is beneficial in various disease conditions. However, it remains unknown whether human AM/AMBP-1 provides any beneficial effects in renal I/R injury. The objective of our study therefore was to determine whether administration of human AM/AMBP-1 can prevent and/or minimize damage in a rat model of renal I/R injury. METHODS: Male adult rats were subjected to renal I/R injury by bilateral renal pedicle clamping with microvascular clips for 60 min followed by reperfusion. Human AM (12 microg/kg BW) and human AMBP-1 (40 microg/kg BW) or vehicle (52 microg/kg BW human albumin) were given intravenously over 30 min immediately following the clip removal (i.e., reperfusion). Rats were allowed to recover for 24 h post-treatment, and blood and renal tissue samples were collected. Plasma levels of AM were measured using a radioimmunoassay specific for rat AM. Plasma AMBP-1 was measured by Western analysis. Renal water content and serum levels of systemic markers of tissue injury were measured. Serum and renal TNF-alpha levels were also assessed. RESULTS: At 24 h after renal I/R injury, plasma levels of AM were significantly increased while plasma AMBP-1 was markedly decreased. Renal water content and systemic markers of tissue injury (e.g., creatinine, BUN, AST, and ALT) were significantly increased following renal I/R injury. Serum and renal TNF-alpha levels were also increased post injury. Administration of human AM/AMBP-1 decreased renal water content, and plasma levels of creatinine, BUN, AST, and ALT. Serum and renal TNF-alpha levels were also significantly decreased after AM/AMBP-1 treatment. CONCLUSION: Treatment with human AM/AMBP-1 in renal I/R injury significantly attenuated organ injury and the inflammatory response. Thus, human AM combined with human AMBP-1 may be developed as a novel treatment for patients with acute renal I/R injury.


Subject(s)
Adrenomedullin/therapeutic use , Complement Factor H/therapeutic use , Kidney Diseases/prevention & control , Reperfusion Injury/prevention & control , Vasodilator Agents/therapeutic use , Adrenomedullin/blood , Alpha-Globulins , Animals , Humans , Kidney/metabolism , Kidney Diseases/blood , Male , Proteins/metabolism , Rats , Reperfusion Injury/blood , Tumor Necrosis Factor-alpha/metabolism , Water/metabolism
20.
Peptides ; 31(5): 872-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20132852

ABSTRACT

Sepsis is a serious complication for patients with obstructive jaundice. Although administration of adrenomedullin (AM) in combination with its binding protein (AMBP-1) is protective after injury, it remains unknown whether AM/AMBP-1 ameliorates sepsis-induced organ injury and mortality in the setting of biliary obstruction. The aim of this study is, therefore, to test the efficacy of human AM/AMBP-1 in a rat model of obstructive jaundice and polymicrobial sepsis. To study this, obstructive jaundice was induced in male adult rats (275-325g) by common bile duct ligation (BDL). One week after BDL, the rats were subjected to sepsis by cecal ligation and puncture (CLP). Plasma levels of AM and AMBP-1 were measured at 20h after CLP. In additional groups of BDL+CLP rats, human AM/AMBP-1 (24/80microg/kg body weight (BW)) or vehicle (i.e., human albumin) was administered intravenously at 5h after CLP. Blood and tissue samples were collected at 20h after CLP for various measurements. To determine the long-term effect of human AM/AMBP-1 after BDL+CLP, the gangrenous cecum was removed at 20h after CLP and 7-day survival was recorded. Our results showed that plasma levels of AM were significantly increased while AMBP-1 levels were markedly decreased after BDL+CLP (n=8, P<0.05). Administration of human AM/AMBP-1 attenuated tissue injury and inflammatory responses after BDL+CLP. Moreover, human AM/AMBP-1 significantly increased the survival rate from 21% (n=14) to 53% (n=15). Thus, human AM/AMBP-1 ameliorates sepsis-induced organ injury and mortality in jaundiced rats. Human AM/AMBP-1 can be further developed as a novel treatment for sepsis in jaundiced patients.


Subject(s)
Adrenomedullin/blood , Adrenomedullin/therapeutic use , Complement Factor H/metabolism , Complement Factor H/therapeutic use , Jaundice, Obstructive/blood , Sepsis/drug therapy , Sepsis/physiopathology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Cecum/surgery , Complement Factor H/administration & dosage , Creatinine/blood , Humans , Interleukin-6/blood , Lactic Acid/blood , Ligation , Male , Rats , Rats, Sprague-Dawley , Sepsis/blood , Tumor Necrosis Factor-alpha/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...