337 results on '"Blom AM"'
Search Results
2. The complement regulator C4b-binding protein (C4BP) interacts with both the C4c and C4dg subfragments of the parent ligand: evidence for synergy in C4BP subsite binding
- Author
-
LEUNG E, BLOM AM, ISENMAN DE, CLEMENZA, Liliana, LEUNG E, BLOM AM, CLEMENZA L, and ISENMAN DE
- Published
- 2006
3. Zinc supplementation inhibits complement activation in AMD
- Author
-
Smailhodzic, D, van Asten, F, Blom, AM, Mohlin, FC, den Hollander, AI, van de Ven, JPH, van Huet, RAC, Groenewoud, JMM, Tian, Y, Berendschot, TTJM, Lechanteur, YTE, Fauser, S, de Bruijn, C, Daha, MR, van der Wilt, Gj, Hyong, CB, Klevering, BJ, Smailhodzic, D, van Asten, F, Blom, AM, Mohlin, FC, den Hollander, AI, van de Ven, JPH, van Huet, RAC, Groenewoud, JMM, Tian, Y, Berendschot, TTJM, Lechanteur, YTE, Fauser, S, de Bruijn, C, Daha, MR, van der Wilt, Gj, Hyong, CB, and Klevering, BJ
- Published
- 2015
4. C4bBinding Protein (C4BP) activates B cells through the CD40 receptor
- Author
-
Brodeur, Sr, Angelini, F, Bacharier, Lb, Blom, Am, Mizoguchi, E, Fujiwara, H, Plebani, Alessandro, Notarangelo, Ld, Dahlback, B, Tsitsikov, E, and Geha, Rs
- Subjects
CD40 receptor ,C4bBinding protein ,B cells - Published
- 2003
5. Plasma bikunin: half-life and tissue uptake
- Author
-
Kaczmarczyk, A, Blom, AM, Alston-Smith, J, Sjoquist, M, Fries, E, Kaczmarczyk, A, Blom, AM, Alston-Smith, J, Sjoquist, M, and Fries, E
- Published
- 2005
6. Effects of filtration rate on the glomerular barrier and clearance of fourdifferently shaped molecules.
- Author
-
Ohlson, M, Sorensson, J, Lindstrom, K, Blom, AM, Fries, E, Haraldsson, B, Ohlson, M, Sorensson, J, Lindstrom, K, Blom, AM, Fries, E, and Haraldsson, B
- Published
- 2001
7. Bikunin--not just a plasma proteinase inhibitor.
- Author
-
Fries, E, Blom, AM, Fries, E, and Blom, AM
- Published
- 2000
8. Binding of Zn(2+) to the plasma protein inter-alpha-inhibitor.
- Author
-
Blom, AM, Thuveson, M, Kilarski, W, Fries, E, Blom, AM, Thuveson, M, Kilarski, W, and Fries, E
- Published
- 1999
9. Structural characterization of inter-alpha-inhibitor. Evidence for an extended shape.
- Author
-
Blom, AM, Morgelin, M, Oyen, M, Jarvet, J, Fries, E, Blom, AM, Morgelin, M, Oyen, M, Jarvet, J, and Fries, E
- Published
- 1999
10. Backcross and partial advanced intercross analysis of nonobese diabetic gene-mediated effects on collagen-induced arthritis reveals an interactive effect by two major loci
- Author
-
Lindqvist, Akb, Martina Johannesson, Johansson, Acm, Nandakumar, Ks, Blom, Am, and Holmdahl, R.
11. CD59 double knockout mice express a CD59ba hybrid fusion protein that mediates insulin secretion.
- Author
-
Ekström A, Villoutreix BO, Halperin J, Renström E, Blom AM, and King BC
- Subjects
- Animals, Mice, Recombinant Fusion Proteins metabolism, Recombinant Fusion Proteins genetics, Blood Glucose metabolism, Insulin-Secreting Cells metabolism, Mice, Inbred C57BL, CD59 Antigens metabolism, CD59 Antigens genetics, Mice, Knockout, Insulin Secretion, Insulin metabolism
- Abstract
CD59 is a cell-surface inhibitor of the terminal step in the complement cascade. However, in addition to its complement inhibitory function, a non-canonical role of CD59 in pancreatic beta cells has been identified. Two recently discovered intracellular alternative splice forms of CD59, IRIS-1 and IRIS-2, are involved in insulin exocytosis through interactions with SNARE-complex components. In mice, the CD59 gene has undergone duplication and to further explore the role of CD59 in insulin secretion, blood glucose homeostasis was studied in a CD59 double knockout (CD59abKO) mouse model. However, no phenotypic deviation related to insulin secretion or blood glucose homeostasis was observed for the CD59abKO mice. Instead, a CD59ba hybrid transcript formed as a consequence of the mutation induced to generate the model was identified. This hybrid transcript is expressed in pancreatic islets of the CD59abKO mice and is comprised of the remaining exons of the two CD59 genes spliced together. Similar to canonical CD59, the CD59ba hybrid was found to be glycosylated and present on the cell surface when exogenously expressed in INS-1 832/13 cells. Furthermore, INS-1 832/13 cells over-expressing the mouse CD59ba hybrid retained normal insulin secretion following siRNA-mediated knockdown of canonical CD59. Hence, although the CD59ba hybrid has lost the complement inhibitory function, the intracellular insulin secretory function remains. These results provide further information concerning the structural requirements of CD59 in its intracellular role relative to its role as a complement inhibitor. It also highlights the importance of carefully assessing plausible consequences of induced mutations in research models., (© 2024 The Author(s). The FASEB Journal published by Wiley Periodicals LLC on behalf of Federation of American Societies for Experimental Biology.)
- Published
- 2024
- Full Text
- View/download PDF
12. Shedding of membrane complement inhibitors CD59 and CD46 into the circulation is associated with poor prognosis in acute coronary syndrome patients: a cohort study.
- Author
-
Zhong B, King B, Waziri H, Yndigegn T, Engelbertsen D, Björkbacka H, Nilsson J, Goncalves I, Blom AM, and Schiopu A
- Subjects
- Humans, Male, Female, Prognosis, Cohort Studies, Aged, Middle Aged, Risk Factors, Acute Coronary Syndrome blood, CD59 Antigens blood, Membrane Cofactor Protein blood
- Abstract
Introduction: The role of the complement inhibitory proteins CD46 and CD59 in the immune response to an acute coronary syndrome (ACS) is unknown. We investigated the relationships between the shedding of CD46 and CD59 into the circulation, reflected by plasma levels of soluble CD46 and CD59, and the risk for post-ACS complications., Methods: We measured plasma sCD46 and sCD59 in a cohort of 546 ACS patients within 24 h after hospital admission, and after 6-weeks in a subgroup of 114 patients. Study outcomes were incident heart failure (HF), major adverse cardiovascular events (MACE) and mortality during a median follow-up period of up to 3.3 years. Echocardiography at 1-year was performed in the follow-up subgroup., Results: Elevated sCD46 and sCD59 were correlated with increased levels of inflammatory mediators and metalloproteinases in plasma, and were associated with increased risk for MACE in Cox proportional hazard models adjusted for cardiovascular risk factors and revascularization [HR 95% CI 1.24 (1.02-1.52), p = 0.034 for sCD46 and 1.18 (1.00-1.38), p = 0.049 for sCD59]. Elevated sCD59 was also associated with higher incidence of HF [HR 95% CI 1.41 (1.15-1.74), p = 0.001], and with lower left ventricular ejection fraction at 1-year post-ACS (Spearman r = - 0.234, p = 0.020). We found no associations between plasma levels of the proteins at 6 weeks and outcomes., Conclusions: Shedding of the complement regulators CD46 and CD59 in plasma in the acute phase of ACS is associated with a negative prognosis. Plasma sCD46 and sCD59 could reflect the degree of local immune activation and serve as prognostic biomarkers in ACS patients., Competing Interests: Declarations Ethics approval and consent to participate All patients signed an informed consent form before inclusion in the study. The study was approved by the Regional Ethics Committee for human studies in Lund, Sweden (reference number 211/2007). Consent for publication Not applicable. Competing interests The authors declare that they have no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. C4b-Binding Protein and Factor H Inhibit Inflammasome Activation during Group A Streptococci Infection in Human Cells.
- Author
-
Bettoni S, Dziedzic M, Bierschenk D, Chrobak M, Magda M, Laabei M, King BC, Riesbeck K, and Blom AM
- Abstract
Introduction: Streptococcus pyogenes (Group A Streptococcus; GAS) is a pathogen that causes over half a million deaths annually worldwide. Human immune cells respond to GAS infection by activating the NLRP3 inflammasome that leads to pro-inflammatory cytokines release which acts to control infection. We investigated the role of C4b-binding protein (C4BP) and Factor H (FH) in the inflammasome response to GAS, as they are recruited by GAS to prevent complement deposition and limit phagocytosis., Methods: Inflammasome response was investigated using isolated primary human cells and the GAS-AP1 strain. Cytokine responses were evaluated by ELISA. C4BP internalisation was investigated using confocal microscopy. Western blotting was used to evaluate the activation of NLRP3 inflammasome components., Results: IL-1β release, induced by GAS-AP1, was inhibited by FH which interferes with priming of human cells. In contrast, C4BP restricted the IL-1β response with no effect on cell priming. C4BP was engulfed by cells together with bacteria and excluded from low-pH vesicles, but localised within the cytosol and near the ASC speck inflammasome complex. C4BP did not inhibit either the inflammasome complex assembly or caspase-1 activation. However, C4BP limited the cleavage of gasderminD N-terminal fragments by interfering with caspase-1 enzymatic activity., Conclusion: Our results provide new insights on the effect of FH and internalised C4BP to control GAS sensing by inflammasomes., (The Author(s). Published by S. Karger AG, Basel.)
- Published
- 2024
- Full Text
- View/download PDF
14. Pandrug-resistant Klebsiella pneumoniae isolated from Ukrainian war victims are hypervirulent.
- Author
-
Ljungquist O, Magda M, Giske CG, Tellapragada C, Nazarchuk O, Dmytriiev D, Thofte O, Öhnström V, Matuschek E, Blom AM, and Riesbeck K
- Abstract
Objectives: Carbapenem- and colistin-resistant Klebsiella pneumoniae were isolated from war victims treated in hospitals in Ukraine. The question was whether these pandrug-resistant K. pneumoniae are pathogenic and capable of causing disease in a broader context., Methods: Klebsiella pneumoniae clinical isolates (n = 37) were tested for antibiotic resistance and subjected to whole-genome sequencing (WGS). In addition, their pathogenicity was tested by serum resistance and two separate animal models., Results: Isolates belonging to the sequence types (ST) 23, 147, 307, 395, and 512 were found to harbor resistance genes against carbapenems and cephalosporins. Nine isolates carried point mutations in pmrB and phoP genes associated with colistin resistance. All bacteria were equipped with multiple virulence genes, and the colistin-resistant isolates each carried 10 different genes. Colistin-resistant K. pneumoniae were more serum-resistant, more virulent against G. mellonella larvae, and displayed an increased survival in mice compared to colistin-susceptible bacteria. The iucA, peg-344, rmpA, rmpC, and rmpD genes were associated with increased virulence in animals., Conclusions: Pandrug-resistant K. pneumoniae in Ukraine are hypervirulent and retain their pathogenicity, highlighting the need to prevent disseminated spread., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
15. Complement factor H is an ICOS ligand modulating Tregs in the glioma microenvironment.
- Author
-
Smolag KI, Olszowka J, Rosberg R, Johansson E, Marinko E, Leandersson K, O'Connell DJ, Governa V, Tuysuz EC, Belting M, Pietras A, Martin M, and Blom AM
- Abstract
The survival rate of glioma patients has not significantly increased in recent years despite aggressive treatment and advances in immunotherapy. The limited response to treatments is partially attributed to the immunosuppressive tumor microenvironment, where regulatory T cells (Tregs) play a pivotal role in immunological tolerance. In this study, we investigated the impact of complement factor H (FH) on Tregs within the glioma microenvironment and found that FH is an ICOS ligand. The binding of FH to this immune checkpoint molecule promoted the survival and function of Tregs and induced the secretion of TGF-beta (TGF-β) and IL-10, while also suppressing T-cell proliferation. We further demonstrated that cancer cells in human and mouse gliomas directly produce FH. Database investigations revealed that upregulation of FH expression was associated with the presence of Tregs and correlated with worse prognosis for glioma patients. We confirmed the effect of FH on glioma development in a mouse model, where FH knockdown was associated with decrease in number of ICOS+ Tregs and demonstrated a tendency of prolonged survival (p=0.064). Since the accumulation of Tregs represents a promising prognostic and therapeutic target, evaluating FH expression should be considered when assessing the effectiveness of and resistance to immunotherapies against glioma.
- Published
- 2024
- Full Text
- View/download PDF
16. Hypoxia-induced complement component 3 promotes aggressive tumor growth in the glioblastoma microenvironment.
- Author
-
Rosberg R, Smolag KI, Sjölund J, Johansson E, Bergelin C, Wahldén J, Pantazopoulou V, Ceberg C, Pietras K, Blom AM, and Pietras A
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Macrophages metabolism, Macrophages immunology, Signal Transduction, Microglia metabolism, Microglia pathology, Disease Models, Animal, Hypoxia metabolism, Arginine analogs & derivatives, Benzhydryl Compounds, Receptors, G-Protein-Coupled, Glioblastoma pathology, Glioblastoma genetics, Glioblastoma metabolism, Tumor Microenvironment immunology, Complement C3 metabolism, Complement C3 genetics, Brain Neoplasms pathology, Brain Neoplasms genetics, Brain Neoplasms metabolism, Receptors, Complement metabolism, Receptors, Complement genetics
- Abstract
Glioblastoma (GBM) is the most aggressive form of glioma with a high rate of relapse despite intensive treatment. Tumor recurrence is tightly linked to radio-resistance, which in turn is associated with hypoxia. Here, we discovered a strong link between hypoxia and local complement signaling using publicly available bulk, single-cell, and spatially resolved transcriptomic data from patients with GBM. Complement component 3 (C3) and the receptor C3AR1 were both associated with aggressive disease and shorter survival in human glioma. In a genetically engineered mouse model of GBM, we found C3 specifically in hypoxic tumor areas. In vitro, we found an oxygen level-dependent increase in C3 and C3AR1 expression in response to hypoxia in several GBM and stromal cell types. C3a induced M2 polarization of cultured microglia and macrophages in a C3aR-dependent fashion. Targeting C3aR using the antagonist SB290157 prolonged survival of glioma-bearing mice both alone and in combination with radiotherapy while reducing the number of M2-polarized macrophages. Our findings establish a strong link between hypoxia and complement pathways in GBM and support a role of hypoxia-induced C3a/C3aR signaling as a contributor to glioma aggressiveness by regulating macrophage polarization.
- Published
- 2024
- Full Text
- View/download PDF
17. Intracellular complement and immunometabolism: The advantages of compartmentalization.
- Author
-
King BC and Blom AM
- Subjects
- Humans, Animals, Phagocytosis immunology, Inflammation immunology, Inflammation metabolism, Complement System Proteins immunology, Complement System Proteins metabolism, Signal Transduction immunology, Complement Activation immunology
- Abstract
The complement system is a proteolytic cascade triggered by pathogen and danger-associated molecular patterns, with resultant outcomes of inflammation, cellular activation, and opsonization of material for removal by phagocytosis. While first discovered as an activity in serum, it is now recognized that complement components play important roles at local and individual cell-intrinsic levels. In particular, apart from the extracellular serum activities of complement, it is now believed that complement also acts intracellularly, as part of a cellular signal transduction cascade that can stimulate cellular survival and activation, and individual immune cell phenotypes, via effects on cellular metabolism. This review will describe what is currently known about how complement functions in intracellular signal transduction, and outline the functional advantages of a compartmentalized and intracellular complement system., (© 2024 The Authors. European Journal of Immunology published by Wiley‐VCH GmbH.)
- Published
- 2024
- Full Text
- View/download PDF
18. Intracellular cartilage oligomeric matrix protein augments breast cancer resistance to chemotherapy.
- Author
-
Hanitrarimalala V, Bednarska I, Murakami T, Papadakos KS, and Blom AM
- Subjects
- Humans, Female, Cell Line, Tumor, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum drug effects, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Breast Neoplasms pathology, Breast Neoplasms genetics, Drug Resistance, Neoplasm drug effects, Cartilage Oligomeric Matrix Protein metabolism, Cartilage Oligomeric Matrix Protein genetics, Apoptosis drug effects, Calpain metabolism
- Abstract
Chemotherapy persists as the primary intervention for breast cancer, with chemoresistance posing the principal obstacle to successful treatment. Herein, we show that cartilage oligomeric matrix protein (COMP) expression leads to increased cancer cell survival and attenuated apoptosis under treatment with several chemotherapeutic drugs, anti-HER2 targeted treatment, and endocrine therapy in several breast cancer cell lines tested. The COMP-induced chemoresistance was independent of the breast cancer subtype. Extracellularly delivered recombinant COMP failed to rescue cells from apoptosis while endoplasmic reticulum (ER)-restricted COMP-KDEL conferred resistance to apoptosis, consistent with the localization of COMP in the ER, where it interacted with calpain. Calpain activation was reduced in COMP-expressing cells and maintained at a lower level of activation during treatment with epirubicin. Moreover, the downstream caspases of calpain, caspases -9, -7, and -3, exhibited significantly reduced activation in COMP-expressing cells under chemotherapy treatment. Chemotherapy, when combined with calpain activators, rendered the cells expressing COMP more chemosensitive. Also, the anti-apoptotic proteins phospho-Bcl2 and survivin were increased in COMP-expressing cells upon chemotherapy. Cells expressing a mutant COMP lacking thrombospondin repeats exhibited reduced chemoresistance compared to cells expressing full-length COMP. Evaluation of calcium levels in the ER, cytosol, and mitochondria revealed that COMP expression modulates intracellular calcium homeostasis. Furthermore, patients undergoing chemotherapy or endocrine therapy demonstrated significantly reduced overall survival time when tumors expressed high levels of COMP. This study identifies a novel role of COMP in chemoresistance and calpain inactivation in breast cancer, a discovery with potential implications for anti-cancer therapy., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
19. CSMD1 regulates brain complement activity and circuit development.
- Author
-
Baum ML, Wilton DK, Fox RG, Carey A, Hsu YH, Hu R, Jäntti HJ, Fahey JB, Muthukumar AK, Salla N, Crotty W, Scott-Hewitt N, Bien E, Sabatini DA, Lanser TB, Frouin A, Gergits F, Håvik B, Gialeli C, Nacu E, Lage K, Blom AM, Eggan K, McCarroll SA, Johnson MB, and Stevens B
- Subjects
- Animals, Humans, Mice, Cells, Cultured, Complement C3 metabolism, Complement System Proteins metabolism, Mice, Inbred C57BL, Microglia metabolism, Thalamus metabolism, Brain metabolism, Membrane Proteins metabolism, Membrane Proteins genetics, Mice, Knockout, Neurons metabolism, Synapses metabolism
- Abstract
Complement proteins facilitate synaptic elimination during neurodevelopmental pruning, but neural complement regulation is not well understood. CUB and Sushi Multiple Domains 1 (CSMD1) can regulate complement activity in vitro, is expressed in the brain, and is associated with increased schizophrenia risk. Beyond this, little is known about CSMD1 including whether it regulates complement activity in the brain or otherwise plays a role in neurodevelopment. We used biochemical, immunohistochemical, and proteomic techniques to examine the regional, cellular, and subcellular distribution as well as protein interactions of CSMD1 in the brain. To evaluate whether CSMD1 is involved in complement-mediated synapse elimination, we examined Csmd1-knockout mice and CSMD1-knockout human stem cell-derived neurons. We interrogated synapse and circuit development of the mouse visual thalamus, a process that involves complement pathway activity. We also quantified complement deposition on synapses in mouse visual thalamus and on cultured human neurons. Finally, we assessed uptake of synaptosomes by cultured microglia. We found that CSMD1 is present at synapses and interacts with complement proteins in the brain. Mice lacking Csmd1 displayed increased levels of complement component C3, an increased colocalization of C3 with presynaptic terminals, fewer retinogeniculate synapses, and aberrant segregation of eye-specific retinal inputs to the visual thalamus during the critical period of complement-dependent refinement of this circuit. Loss of CSMD1 in vivo enhanced synaptosome engulfment by microglia in vitro, and this effect was dependent on activity of the microglial complement receptor, CR3. Finally, human stem cell-derived neurons lacking CSMD1 were more vulnerable to complement deposition. These data suggest that CSMD1 can function as a regulator of complement-mediated synapse elimination in the brain during development., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
20. Elevated expression of complement factor I in lung cancer cells associates with shorter survival-Potentially via non-canonical mechanism.
- Author
-
Felberg A, Bieńkowski M, Stokowy T, Myszczyński K, Polakiewicz Z, Kitowska K, Sądej R, Mohlin F, Kuźniewska A, Kowalska D, Stasiłojć G, Jongerius I, Spaapen R, Mesa-Guzman M, Montuenga LM, Blom AM, Pio R, and Okrój M
- Subjects
- Humans, Male, Female, Middle Aged, Cell Line, Tumor, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung mortality, Aged, Prognosis, Gene Expression Regulation, Neoplastic, Lung Neoplasms pathology, Lung Neoplasms metabolism, Lung Neoplasms mortality, Lung Neoplasms genetics, Complement Factor I metabolism, Complement Factor I genetics
- Abstract
While numerous membrane-bound complement inhibitors protect the body's cells from innate immunity's autoaggression, soluble inhibitors like complement factor I (FI) are rarely produced outside the liver. Previously, we reported the expression of FI in non-small cell lung cancer (NSCLC) cell lines. Now, we assessed the content of FI in cancer biopsies from lung cancer patients and associated the results with clinicopathological characteristics and clinical outcomes. Immunohistochemical staining intensity did not correlate with age, smoking status, tumor size, stage, differentiation grade, and T cell infiltrates, but was associated with progression-free survival (PFS), overall survival (OS) and disease-specific survival (DSS). Multivariate Cox analysis of low vs. high FI content revealed HR 0.55, 95 % CI 0.32-0.95, p=0.031 for PFS, HR 0.51, 95 % CI 0.25-1.02, p=0.055 for OS, and HR 0.32, 95 % CI 0.12-0.84, p=0.021 for DSS. Unfavorable prognosis might stem from the non-canonical role of FI, as the staining pattern did not correlate with C4d - the product of FI-supported degradation of active complement component C4b. To elucidate that, we engineered three human NSCLC cell lines naturally expressing FI with CRISPR/Cas9 technology, and compared the transcriptome of FI-deficient and FI-sufficient clones in each cell line. RNA sequencing revealed differentially expressed genes engaged in intracellular signaling pathways controlling proliferation, apoptosis, and responsiveness to growth factors. Moreover, in vitro colony-formation assays showed that FI-deficient cells formed smaller foci than FI-sufficient NSCLC cells, but their size increased when purified FI protein was added to the medium. We postulate that a non-canonical activity of FI influences cellular physiology and contributes to the poor prognosis of lung cancer patients., Competing Interests: Declaration of Competing Interest MO and AMB are named as inventors in patent WO2016038055A1 for the anti-C4d antibody used in this study. The other authors declare no conflict of interest. All authors have read the journal's policy on disclosure of potential conflicts of interest., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
21. Identification of an osteopontin-derived peptide that binds neuropilin-1 and activates vascular repair responses and angiogenesis.
- Author
-
Chen Y, Gialeli C, Shen J, Dunér P, Walse B, Duelli A, Caing-Carlsson R, Blom AM, Zibert JR, Nilsson AH, Alenfall J, Liang C, and Nilsson J
- Subjects
- Humans, Animals, Cell Movement drug effects, Vascular Endothelial Growth Factor Receptor-2 metabolism, Cell Proliferation drug effects, Myocytes, Smooth Muscle drug effects, Myocytes, Smooth Muscle metabolism, Male, Peptides pharmacology, Vascular Endothelial Growth Factor A metabolism, Apoptosis drug effects, Mice, Inbred C57BL, Protein Binding, Ischemia drug therapy, Ischemia metabolism, Mice, Angiogenesis, Neuropilin-1 metabolism, Human Umbilical Vein Endothelial Cells drug effects, Neovascularization, Physiologic drug effects, Osteopontin metabolism, Osteopontin genetics
- Abstract
The osteopontin-derived peptide FOL-005 stimulates hair growth. Using ligand-receptor glyco-capture technology we identified neuropilin-1 (NRP-1), a known co-receptor for vascular endothelial growth factor (VEGF) receptors, as the most probable receptor for FOL-005 and the more stable analogue FOL-026. X-ray diffraction and microscale thermophoresis analysis revealed that FOL-026 shares binding site with VEGF in the NRP-1 b1-subdomain. Stimulation of human umbilical vein endothelial cells with FOL-026 resulted in phosphorylation of VEGFR-2, ERK1/2 and AKT, increased cell growth and migration, stimulation of endothelial tube formation and inhibition of apoptosis in vitro. FOL-026 also promoted angiogenesis in vivo as assessed by subcutaneous Matrigel plug and hind limb ischemia models. NRP-1 knock-down or treatment of NRP-1 antagonist EG00229 blocked the stimulatory effects of FOL-026 on endothelial cells. Exposure of human coronary artery smooth muscle cells to FOL-026 stimulated cell growth, migration, inhibited apoptosis, and induced VEGF gene expression and VEGFR-2/AKT phosphorylation by an NRP-1-dependent mechanism. RNA sequencing showed that FOL-026 activated pathways involved in tissue repair. These findings identify NRP-1 as the receptor for FOL-026 and show that its biological effects mimic that of growth factors binding to the VEGF receptor family. They also suggest that FOL-026 may have therapeutical potential in conditions that require vascular repair and/or enhanced angiogenesis., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Jan Nilsson reports financial support was provided by Novo Nordisk Foundation. Yihong Chen reports financial support was provided by Shanghai Sailing Program. Chun Liang reports financial support was provided by National Natural Science Foundation of China. Jan Nilsson reports a relationship with Coegin Pharma that includes: consulting or advisory, equity or stocks, and funding grants. Anna Hultgaardh reports a relationship with Coegin Pharma that includes: consulting or advisory and equity or stocks. Jan Nilsson has patent issued to Coegin Pharma. Anna Hultgaardh has patent pending to Coegin Pharma. None If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. Pontus Duner has stocks in Coegin Pharma., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
22. Targeted complement inhibition using bispecific antibodies that bind local antigens and endogenous complement regulators.
- Author
-
Wang H, van de Bovenkamp FS, Dijkstra DJ, Abendstein L, Borggreven NV, Pool J, Zuijderduijn R, Gstöttner C, Gelderman KA, Damelang T, Vidarsson G, Blom AM, Domínguez-Vega E, Parren PWHI, Sharp TH, and Trouw LA
- Subjects
- Humans, Antigens immunology, Complement System Proteins immunology, Complement System Proteins metabolism, Protein Binding, Antibodies, Bispecific immunology, Antibodies, Bispecific pharmacology, Complement Activation immunology, Complement C4b-Binding Protein immunology, Complement C4b-Binding Protein metabolism, Complement Factor H immunology, Complement Factor H metabolism
- 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., Competing Interests: Author PWHIP was employed by Gyes BV. FSB, PWHIP, and LAT are listed as inventors on a patent application regarding the application of targeted complement inhibition. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Wang, van de Bovenkamp, Dijkstra, Abendstein, Borggreven, Pool, Zuijderduijn, Gstöttner, Gelderman, Damelang, Vidarsson, Blom, Domínguez-Vega, Parren, Sharp and Trouw.)
- Published
- 2024
- Full Text
- View/download PDF
23. Stromal cartilage oligomeric matrix protein as a tumorigenic driver in ovarian cancer via Notch3 signaling and epithelial-to-mesenchymal transition.
- Author
-
Gorji-Bahri G, Krishna BM, Hagerling C, Orimo A, Jirström K, Papadakos KS, and Blom AM
- Subjects
- Humans, Animals, Mice, Female, Cartilage Oligomeric Matrix Protein, Receptor, Notch3, Carcinogenesis, Signal Transduction, Ovarian Neoplasms
- Abstract
Background: Cartilage oligomeric matrix protein (COMP), an extracellular matrix glycoprotein, is vital in preserving cartilage integrity. Further, its overexpression is associated with the aggressiveness of several types of solid cancers. This study investigated COMP's role in ovarian cancer, exploring clinicopathological links and mechanistic insights., Methods: To study the association of COMP expression in cancer cells and stroma with clinicopathological features of ovarian tumor patients, we analyzed an epithelial ovarian tumor cohort by immunohistochemical analysis. Subsequently, to study the functional mechanisms played by COMP, an in vivo xenograft mouse model and several molecular biology techniques such as transwell migration and invasion assay, tumorsphere formation assay, proximity ligation assay, and RT-qPCR array were performed., Results: Based on immunohistochemical analysis of epithelial ovarian tumor tissues, COMP expression in the stroma, but not in cancer cells, was linked to worse overall survival (OS) of ovarian cancer patients. A xenograft mouse model showed that carcinoma-associated fibroblasts (CAFs) expressing COMP stimulate the growth and metastasis of ovarian tumors through the secretion of COMP. The expression of COMP was upregulated in CAFs stimulated with TGF-β. Functionally, secreted COMP by CAFs enhanced the migratory capacity of ovarian cancer cells. Mechanistically, COMP activated the Notch3 receptor by enhancing the Notch3-Jagged1 interaction. The dependency of the COMP effect on Notch was confirmed when the migration and tumorsphere formation of COMP-treated ovarian cancer cells were inhibited upon incubation with Notch inhibitors. Moreover, COMP treatment induced epithelial-to-mesenchymal transition and upregulation of active β-catenin in ovarian cancer cells., Conclusion: This study suggests that COMP secretion by CAFs drives ovarian cancer progression through the induction of the Notch pathway and epithelial-to-mesenchymal transition., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
24. The prognostic and potentially immunomodulatory role of cartilage oligomeric matrix protein in patients with gastric and esophageal adenocarcinoma.
- Author
-
Papadakos KS, Gorji-Bahri G, Gialeli C, Hedner C, Hagerling C, Svensson MC, Jeremiasen M, Borg D, Fristedt R, Jirström K, and Blom AM
- Subjects
- Humans, Cartilage Oligomeric Matrix Protein, Prognosis, Collagen, Tumor Microenvironment, Adenocarcinoma, Stomach Neoplasms, Esophageal Neoplasms
- Abstract
Background: Cartilage oligomeric matrix protein (COMP) is a novel regulator of the tumor microenvironment. Studies in colon cancer and pancreatobiliary adenocarcinoma have revealed COMP expression to be associated with decreased infiltration of immune cells in the tumor microenvironment. Herein, the expression of COMP was investigated in gastric and esophageal adenocarcinoma with particular reference to its the relationship with the immune microenvironment., Methods: COMP expression was evaluated in tissue microarrays representing primary tumors from 159 patients with chemo- and radiotherapy naïve esophageal and gastric adenocarcinoma and 67 matched samples of lymph node metastases using immunohistochemistry. Additionally, collagen fibers were stained with Sirius Red and evaluated with the FIJI macro TWOMBLI algorithm., Results: The expression of COMP in cancer cells in the entire cohort was associated with shorter overall survival (OS) (p = 0.013) and recurrence-free survival (RFS) (p = 0.029), while COMP expression in the stroma was correlated with shorter RFS (p = 0.042). Similar correlations were found for patients with gastric adenocarcinoma, whereas COMP expression was not prognostic in esophageal adenocarcinoma. Further, in the entire cohort, the expression of COMP in the stroma was correlated with exclusion of different populations of immune cells (CD8
+ , CD3+ , FoxP3+ , CD20+ ) from the tumor microenvironment. Finally, higher density and alignment of collagen fibers were correlated with the expression of COMP in the stroma., Conclusions: Expression of COMP in gastric and esophageal adenocarcinoma was correlated with shorter OS and RFS. A reduced number of immune cells infiltrated the tumor microenvironment when COMP expression was detected. This phenomenon could be attributed to the denser collagen deposits, a hallmark of tumor fibrosis observed in COMP-expressing tumors., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
25. Tumor suppressor role of the complement inhibitor CSMD1 and its role in TNF-induced neuroinflammation in gliomas.
- Author
-
Tuysuz EC, Mourati E, Rosberg R, Moskal A, Gialeli C, Johansson E, Governa V, Belting M, Pietras A, and Blom AM
- Subjects
- Humans, Animals, Mice, Neuroinflammatory Diseases, Proto-Oncogene Proteins c-sis genetics, Disease-Free Survival, Isocitrate Dehydrogenase genetics, Mutation, Membrane Proteins genetics, Tumor Suppressor Proteins genetics, Glioma pathology, Brain Neoplasms pathology
- Abstract
Background: The complement inhibitor CSMD1 acts as a tumor suppressor in various types of solid cancers. Despite its high level of expression in the brain, its function in gliomas, malignant brain tumors originating from glial cells, has not been investigated., Methods: Three cohorts of glioma patients comprising 1500 patients were analyzed in our study along with their clinical data. H4, U-118 and U-87 cell lines were used to investigate the tumor suppressor function of CSMD1 in gliomas. PDGFB-induced brain tumor model was utilized for the validation of in vitro data., Results: The downregulation of CSMD1 expression correlated with reduced overall and disease-free survival, elevated tumor grade, wild-type IDH genotype, and intact 1p/19q status. Moreover, enhanced activity was noted in the neuroinflammation pathway. Importantly, ectopic expression of CSMD1 in glioma cell lines led to decreased aggressiveness in vitro. Mechanically, CSMD1 obstructed the TNF-induced NF-kB and STAT3 signaling pathways, effectively suppressing the secretion of IL-6 and IL-8. There was also reduced survival in PDGFB-induced brain tumors in mice when Csmd1 was downregulated., Conclusions: Our study has identified CSMD1 as a tumor suppressor in gliomas and elucidated its role in TNF-induced neuroinflammation, contributing to a deeper understanding of glioma pathogenesis., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
26. Intracellular C3 protects β-cells from IL-1β-driven cytotoxicity via interaction with Fyn-related kinase.
- Author
-
Kulak K, Kuska K, Colineau L, Mckay M, Maziarz K, Slaby J, Blom AM, and King BC
- Subjects
- Mice, Animals, Humans, Cell Death, Cytokines metabolism, Mice, Knockout, Diabetes Mellitus, Type 2 metabolism, Insulin-Secreting Cells metabolism, Islets of Langerhans metabolism
- Abstract
One of the hallmarks of type 1 but also type 2 diabetes is pancreatic islet inflammation, associated with altered pancreatic islet function and structure, if unresolved. IL-1β is a proinflammatory cytokine which detrimentally affects β-cell function. In the course of diabetes, complement components, including the central complement protein C3, are deregulated. Previously, we reported high C3 expression in human pancreatic islets, with upregulation after IL-1β treatment. In the current investigation, using primary human and rodent material and CRISPR/Cas9 gene-edited β-cells deficient in C3, or producing only cytosolic C3 from a noncanonical in-frame start codon, we report a protective effect of C3 against IL-1β-induced β-cell death, that is attributed to the cytosolic fraction of C3. Further investigation revealed that intracellular C3 alleviates IL-1β-induced β-cell death, by interaction with and inhibition of Fyn-related kinase (FRK), which is involved in the response of β-cells to cytokines. Furthermore, these data were supported by increased β-cell death in vivo in a β-cell-specific C3 knockout mouse. Our data indicate that a functional, cytoprotective association exists between FRK and cytosolic C3., Competing Interests: Competing interests statement:The authors declare no competing interest.
- Published
- 2024
- Full Text
- View/download PDF
27. Complement activation negatively affects the platelet response to thrombopoietin receptor agonists in patients with immune thrombocytopenia: a prospective cohort study.
- Author
-
Åkesson A, Bussel JB, Martin M, Blom AM, Klintman J, Ghanima W, Zetterberg E, and Garabet L
- Subjects
- Humans, Receptors, Thrombopoietin agonists, Prospective Studies, Biomarkers, Complement Activation, Thrombopoietin pharmacology, Thrombopoietin therapeutic use, Recombinant Fusion Proteins, Purpura, Thrombocytopenic, Idiopathic, Thrombocytopenia
- Abstract
Increased platelet destruction is central in the pathogenesis of immune thrombocytopenia. However, impaired platelet production is also relevant and its significance underlies the rationale for treatment with thrombopoietin receptor agonists (TPO-RAs). Previous studies have associated enhanced complement activation with increased disease severity. Additionally, treatment refractoriness has been demonstrated to resolve by the administration of complement-targeted therapeutics in a subset of patients. The association between complement activation and the platelet response to TPO-RA therapy has previously not been investigated. In this study, blood samples from patients with immune thrombocytopenia (n = 15) were prospectively collected before and two, six and 12 weeks after the initiation of TPO-RA therapy. Plasma levels of complement degradation product C4d and soluble terminal complement complexes were assessed. Patients with significantly elevated baseline levels of terminal complement complexes exhibited more often an inadequate platelet response ( p = .04), were exclusively subjected to rescue therapy with intravenous immunoglobulin ( p = .02), and did not respond with a significant platelet count increase during the study period. C4d showed a significant ( p = .01) ability to distinguish samples with significant terminal complement activation, implying engagement of the classical complement pathway. In conclusion, elevated levels of complement biomarkers were associated with a worse TPO-RA treatment response. Larger studies are needed to confirm these results. Biomarkers of complement activation may prove valuable as a prognostic tool to predict which patients that potentially could benefit from complement-inhibiting therapy in the future.
- Published
- 2023
- Full Text
- View/download PDF
28. C4BP(β-)-mediated immunomodulation attenuates inflammation in DSS-induced murine colitis and in myeloid cells from IBD patients.
- Author
-
Serrano I, Luque A, Ruiz-Cerulla A, Navas S, Blom AM, Rodríguez de Córdoba S, Fernández FJ, Cristina Vega M, Rodríguez-Moranta F, Guardiola J, and Aran JM
- Subjects
- Animals, Humans, Mice, Anti-Inflammatory Agents pharmacology, Anti-Inflammatory Agents therapeutic use, Immunomodulation, Inflammation, Myeloid Cells, Colitis chemically induced, Colitis drug therapy, Inflammatory Bowel Diseases chemically induced, Inflammatory Bowel Diseases drug therapy
- Abstract
The most recent and promising therapeutic strategies for inflammatory bowel disease (IBD) have engaged biologics targeting single effector components involved in major steps of the immune-inflammatory processes, such as tumor necrosis factor, interleukins or integrins. Nevertheless, these molecules have not yet met expectations regarding efficacy and safety, resulting in a significant percentage of refractory or relapsing patients. Thus, novel treatment options are urgently needed. The minor isoform of the complement inhibitor C4b-binding protein, C4BP(β-), has been shown to confer a robust anti-inflammatory and immunomodulatory phenotype over inflammatory myeloid cells. Here we show that C4BP(β-)-mediated immunomodulation can significantly attenuate the histopathological traits and preserve the intestinal epithelial integrity in dextran sulfate sodium (DSS)-induced murine colitis. C4BP(β-) downregulated inflammatory transcripts, notably those related to neutrophil activity, mitigated circulating inflammatory effector cytokines and chemokines such as CXCL13, key in generating ectopic lymphoid structures, and, overall, prevented inflammatory immune cell infiltration in the colon of colitic mice. PRP6-HO7, a recombinant curtailed analogue with only immunomodulatory activity, achieved a similar outcome as C4BP(β-), indicating that the therapeutic effect is not due to the complement inhibitory activity. Furthermore, both C4BP(β-) and PRP6-HO7 significantly reduced, with comparable efficacy, the intrinsic and TLR-induced inflammatory markers in myeloid cells from both ulcerative colitis and Crohn's disease patients, regardless of their medication. Thus, the pleiotropic anti-inflammatory and immunomodulatory activity of PRP6-HO7, able to "reprogram" myeloid cells from the complex inflammatory bowel environment and to restore immune homeostasis, might constitute a promising therapeutic option for IBD., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Josep M. Aran reports financial support was provided by Ministerio de Ciencia, Innovación y Universidades. Josep M. Aran reports financial support was provided by Departament de Recerca i Universitats de la Generalitat de Catalunya. Josep M. Aran reports financial support was provided by La Caixa Foundation. Josep M. Aran reports financial support was provided by The hna Foundation. Josep M. Aran has patent pending to IDIBELL. Inmaculada Serrano has patent pending to IDIBELL. Ana Luque has patent pending to IDIBELL., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
29. Prompt Thrombo-Inflammatory Response to Ischemia-Reperfusion Injury and Kidney Transplant Outcomes.
- Author
-
Strandberg G, Öberg CM, Blom AM, Slivca O, Berglund D, Segelmark M, Nilsson B, and Biglarnia AR
- Abstract
Introduction: In kidney transplantation (KT), the role of the intravascular innate immune system (IIIS) in response to ischemia-reperfusion injury (IRI) is not well-understood. Here, we studied parallel changes in the generation of key activation products of the proteolytic cascade systems of the IIIS following living donor (LD) and deceased donor (DD) transplantation and evaluated potential associations with clinical outcomes., Methods: In a cohort study, 63 patients undergoing LD ( n = 26) and DD ( n = 37) transplantation were prospectively included. Fifteen DD kidneys were preserved with hypothermic machine perfusion (HMP), and the remaining were cold stored. Activation products of the kallikrein-kinin, coagulation, and complement systems were measured in blood samples obtained systemically at baseline and locally from the transplant renal vein at 1, 10, and 30 minutes after reperfusion., Results: DD kidneys exhibited a prompt and interlinked activation of all 3 cascade systems of IIIS postreperfusion, indicating a robust and local thrombo-inflammatory response to IRI. In this initial response, the complement activation product sC5b-9 exhibited a robust correlation with other IIIS activation markers and displayed a strong association with short-term and mid-term (24-month) graft dysfunction. In contrast, LD kidneys did not exhibit this thrombo-inflammatory response. The use of HMP was associated with reduced thromboinflammation and preserved mid-term kidney function., Conclusion: Kidneys from DD are vulnerable to a prompt thrombo-inflammatory response to IRI, which adversely affects both short-term and long-term allograft function. Strategies aimed at minimizing graft immunogenicity prior to reperfusion are crucial to mitigate the intricate inflammatory response to IRI., (© 2023 International Society of Nephrology. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
30. A subset of type-II collagen-binding antibodies prevents experimental arthritis by inhibiting FCGR3 signaling in neutrophils.
- Author
-
Xu Z, Xu B, Lundström SL, Moreno-Giró À, Zhao D, Martin M, Lönnblom E, Li Q, Krämer A, Ge C, Cheng L, Liang B, Tong D, Stawikowska R, Blom AM, Fields GB, Zubarev RA, and Holmdahl R
- Subjects
- Humans, Animals, Mice, Neutrophils, Collagen, Autoantibodies, Epitopes, Arthritis, Experimental prevention & control
- Abstract
Rheumatoid arthritis (RA) involves several classes of pathogenic autoantibodies, some of which react with type-II collagen (COL2) in articular cartilage. We previously described a subset of COL2 antibodies targeting the F4 epitope (ERGLKGHRGFT) that could be regulatory. Here, using phage display, we developed recombinant antibodies against this epitope and examined the underlying mechanism of action. One of these antibodies, R69-4, protected against cartilage antibody- and collagen-induced arthritis in mice, but not autoimmune disease models independent of arthritogenic autoantibodies. R69-4 was further shown to cross-react with a large range of proteins within the inflamed synovial fluid, such as the complement protein C1q. Complexed R69-4 inhibited neutrophil FCGR3 signaling, thereby impairing downstream IL-1β secretion and neutrophil self-orchestrated recruitment. Likewise, human isotypes of R69-4 protected against arthritis with comparable efficiency. We conclude that R69-4 abrogates autoantibody-mediated arthritis mainly by hindering FCGR3 signaling, highlighting its potential clinical utility in acute RA., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
31. Recruitment of C4b-binding protein is not a complement evasion strategy employed by Staphylococcus aureus .
- Author
-
Li S, Bettoni S, Mohlin F, Geoghegan JA, Blom AM, and Laabei M
- Subjects
- Humans, Staphylococcus aureus genetics, Complement System Proteins, Staphylococcus, Membrane Proteins, Complement C4b-Binding Protein, Staphylococcal Infections
- Abstract
Complement offers a first line of defence against infection through the opsonization of microbial pathogens, recruitment of professional phagocytes to the infection site and the coordination of inflammatory responses required for the resolution of infection. Staphylococcus aureus is a successful pathogen that has developed multiple mechanisms to thwart host immune responses. Understanding the precise strategies employed by S. aureus to bypass host immunity will be paramount for the development of vaccines and or immunotherapies designed to prevent or limit infection. To gain a better insight into the specific immune evasion mechanisms used by S. aureus we examined the pathogen's interaction with the soluble complement inhibitor, C4b-binding protein (C4BP). Previous studies indicated that S. aureus recruits C4BP using a specific cell-wall-anchored surface protein and that bound C4BP limits complement deposition on the staphylococcal surface. Using flow-cytometric-based bacterial-protein binding assays we observed no interaction between S. aureus and C4BP. Moreover, we offer a precautionary warning that C4BP isolated from plasma can be co-purified with minute quantities of human IgG, which can distort binding analysis between S. aureus and human-derived proteins. Combined our data indicates that recruitment of C4BP is not a complement evasion strategy employed by S. aureus .
- Published
- 2023
- Full Text
- View/download PDF
32. Citrullination of C1-inhibitor as a mechanism of impaired complement regulation in rheumatoid arthritis.
- Author
-
Martin M, Nilsson SC, Eikrem D, Fromell K, Scavenius C, Vogt LM, Bielecka E, Potempa J, Enghild JJ, Nilsson B, Ekdahl KN, Kapetanovic MC, and Blom AM
- Subjects
- Humans, Protein-Arginine Deiminases genetics, Factor XIIa metabolism, Plasma Kallikrein metabolism, Factor XIa, Proteins metabolism, Autoantibodies, Citrullination, Arthritis, Rheumatoid
- Abstract
Background: Dysregulated complement activation, increased protein citrullination, and production of autoantibodies against citrullinated proteins are hallmarks of rheumatoid arthritis (RA). Citrullination is induced by immune cell-derived peptidyl-Arg deiminases (PADs), which are overactivated in the inflamed synovium. We characterized the effect of PAD2- and PAD4-induced citrullination on the ability of the plasma-derived serpin C1-inhibitor (C1-INH) to inhibit complement and contact system activation., Methods: Citrullination of the C1-INH was confirmed by ELISA and Western blotting using a biotinylated phenylglyoxal probe. C1-INH-mediated inhibition of complement activation was analyzed by C1-esterase activity assay. Downstream inhibition of complement was studied by C4b deposition on heat-aggregated IgGs by ELISA, using pooled normal human serum as a complement source. Inhibition of the contact system was investigated by chromogenic activity assays for factor XIIa, plasma kallikrein, and factor XIa. In addition, autoantibody reactivity to native and citrullinated C1-INH was measured by ELISA in 101 RA patient samples., Results: C1-INH was efficiently citrullinated by PAD2 and PAD4. Citrullinated C1-INH was not able to bind the serine protease C1s and inhibit its activity. Citrullination of the C1-INH abrogated its ability to dissociate the C1-complex and thus inhibit complement activation. Consequently, citrullinated C1-INH had a decreased capacity to inhibit C4b deposition via the classical and lectin pathways. The inhibitory effect of C1-INH on the contact system components factor XIIa, plasma kallikrein, and factor XIa was also strongly reduced by citrullination. In RA patient samples, autoantibody binding to PAD2- and PAD4-citrullinated C1-INH was detected. Significantly more binding was observed in anti-citrullinated protein antibody (ACPA)-positive than in ACPA-negative samples., Conclusion: Citrullination of the C1-INH by recombinant human PAD2 and PAD4 enzymes impaired its ability to inhibit the complement and contact systems in vitro . Citrullination seems to render C1-INH more immunogenic, and citrullinated C1-INH might thus be an additional target of the autoantibody response observed in RA patients., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Martin, Nilsson, Eikrem, Fromell, Scavenius, Vogt, Bielecka, Potempa, Enghild, Nilsson, Ekdahl, Kapetanovic and Blom.)
- Published
- 2023
- Full Text
- View/download PDF
33. Correction: Complement inhibitor CSMD1 modulates epidermal growth factor receptor oncogenic signaling and sensitizes breast cancer cells to chemotherapy.
- Author
-
Gialeli C, Tuysuz EC, Staaf J, Guleed S, Paciorek V, Mörgelin M, Papadakos KS, and Blom AM
- Published
- 2023
- Full Text
- View/download PDF
34. C4b-binding protein inhibits particulate- and crystalline-induced NLRP3 inflammasome activation.
- Author
-
Bierschenk D, Papac-Milicevic N, Bresch IP, Kovacic V, Bettoni S, Dziedzic M, Wetsel RA, Eschenburg S, Binder CJ, Blom AM, and King BC
- Subjects
- Animals, Humans, Mice, Complement C4b-Binding Protein metabolism, Macrophages metabolism, Silicon Dioxide pharmacology, Inflammasomes metabolism, NLR Family, Pyrin Domain-Containing 3 Protein genetics, NLR Family, Pyrin Domain-Containing 3 Protein metabolism
- Abstract
Dysregulated NLRP3 inflammasome activation drives a wide variety of diseases, while endogenous inhibition of this pathway is poorly characterised. The serum protein C4b-binding protein (C4BP) is a well-established inhibitor of complement with emerging functions as an endogenously expressed inhibitor of the NLRP3 inflammasome signalling pathway. Here, we identified that C4BP purified from human plasma is an inhibitor of crystalline- (monosodium urate, MSU) and particulate-induced (silica) NLRP3 inflammasome activation. Using a C4BP mutant panel, we identified that C4BP bound these particles via specific protein domains located on the C4BP α-chain. Plasma-purified C4BP was internalised into MSU- or silica-stimulated human primary macrophages, and inhibited MSU- or silica-induced inflammasome complex assembly and IL-1β cytokine secretion. While internalised C4BP in MSU or silica-stimulated human macrophages was in close proximity to the inflammasome adaptor protein ASC, C4BP had no direct effect on ASC polymerisation in in vitro assays. C4BP was also protective against MSU- and silica-induced lysosomal membrane damage. We further provide evidence for an anti-inflammatory function for C4BP in vivo , as C4bp
-/- mice showed an elevated pro-inflammatory state following intraperitoneal delivery of MSU. Therefore, internalised C4BP is an inhibitor of crystal- or particle-induced inflammasome responses in human primary macrophages, while murine C4BP protects against an enhanced inflammatory state in vivo . Our data suggests C4BP has important functions in retaining tissue homeostasis in both human and mice as an endogenous serum inhibitor of particulate-stimulated inflammasome activation., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Bierschenk, Papac-Milicevic, Bresch, Kovacic, Bettoni, Dziedzic, Wetsel, Eschenburg, Binder, Blom and King.)- Published
- 2023
- Full Text
- View/download PDF
35. Correction: Complement inhibitor CSMD1 acts as tumor suppressor in human breast cancer.
- Author
-
Escudero-Esparza A, Bartoschek M, Gialeli C, Okroj M, Owen S, Jirström K, Orimo A, Jiang WG, Pietras K, and Blom AM
- Published
- 2023
- Full Text
- View/download PDF
36. Expression of Cartilage Oligomeric Matrix Protein in colorectal cancer is an adverse prognostic factor and correlates negatively with infiltrating immune cells and PD-L1 expression.
- Author
-
Blom AM, Gialeli C, Hagerling C, Berntsson J, Jirström K, and Papadakos KS
- Subjects
- Humans, Biomarkers, Prognosis, B7-H1 Antigen metabolism, Cartilage Oligomeric Matrix Protein metabolism, Colorectal Neoplasms
- Abstract
Introduction: Cartilage Oligomeric Matrix Protein (COMP) is an oncogenic protein that has been associated with a decrease in infiltrating T-cells in periampullary adenocarcinoma. This study aimed to investigate whether this is also the case for colorectal cancer (CRC) and to evaluate the relationship between COMP expression and clinopathological features., Methods: Immunohistochemistry was used to determine the expression levels of COMP in tumor cells and stroma in primary tumors from a cohort of 537 CRC patients. The expression of immune cell markers, including CD3+, CD8+, FoxP3+, CD68+, CD56+, CD163+, and PD-L1, was evaluated previously. Tumor fibrosis was assessed by Sirius Red staining and evaluation of collagen fiber organization., Results: COMP expression correlated positively with TNM-stage and grade of differentiation. Patients with CRC expressing high levels of COMP had significantly shorter OS than those with low COMP expression (p<0.0001), and fewer infiltrating T-cells were detected in tumors with high COMP expression. Additionally, a negative correlation was identified between the expression of COMP and PD-L1 on both tumor cells and immune cells. Cox regression analysis showed that tumors expressing high levels of COMP had significantly shorter OS, independent of all evaluated immune cell markers. Tumor fibrosis was correlated with high expression of COMP in the stroma (p<0.0001), and tumors with high levels of COMP expression and denser fibrosis displayed more sparse immune cell infiltration., Discussion: The results suggest that COMP expression in CRC may exert an immune regulatory effect by increasing dense fibrosis and decreasing immune cell infiltration. These findings support the notion that COMP is an important factor in the development and progression of CRC., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Blom, Gialeli, Hagerling, Berntsson, Jirström and Papadakos.)
- Published
- 2023
- Full Text
- View/download PDF
37. The complement receptor C3AR constitutes a novel therapeutic target in NPM1-mutated AML.
- Author
-
von Palffy S, Thorsson H, Peña-Martínez P, Puente-Moncada N, Sandén C, Blom AM, Henningsson R, Juliusson G, King B, Landberg N, Lazarevic V, Orsmark-Pietras C, Rissler M, Rissler V, Ågerstam H, Järås M, Lilljebjörn H, and Fioretos T
- Subjects
- Mice, Animals, Nucleophosmin, Signal Transduction, Antigens, CD34, Receptors, G-Protein-Coupled, Nuclear Proteins genetics, Nuclear Proteins metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute metabolism
- Abstract
Mutated nucleophosmin 1 (NPM1) is the most common genetic alteration in acute myeloid leukemia (AML), found in ∼30% of cases. Although mutations in this gene are considered favorable according to current risk stratification guidelines, a large fraction of patients will experience relapse, demonstrating the urgent need for new treatment options. Therefore, we aimed to identify cell surface proteins specifically expressed on NPM1-mutated AML cells, allowing for potential targeting with antibody-based therapies. Herein, we report on an arrayed flow cytometry-based screen directed to 362 cell surface markers. In comparing the cell surface expression on NPM1-mutated AML cells with primitive (CD34+ CD38-) normal bone marrow cells, we identified the complement receptor C3AR as being specifically expressed in NPM1-mutated AML. By flow cytometry and single-cell RNA sequencing, we further show that normal hematopoietic stem and progenitor cells lack detectable C3AR gene and protein expression, making it particularly suitable as a target for antibody therapy. We also demonstrate that C3AR in combination with GPR56 distinguishes the leukemic stem cells (LSCs) in NPM1-mutated AML from the normal hematopoietic stem cells, defining the LSC population, as shown by transplantation into immunodeficient mice. Mechanistically, the stimulation of C3AR-expressing cells with C3a, the ligand of C3AR, leads to the activation of ERK1/2 and increased survival of AML cells, suggesting that this is an important signaling axis in this subtype of AML. Finally, we show that antibodies directed against C3AR efficiently elicit natural killer cell-mediated killing of primary AML cells ex vivo, highlighting C3AR as a candidate therapeutic target in NPM1-mutated AML., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
38. Neisseria gonorrhoeae co-opts C4b-binding protein to enhance complement-independent survival from neutrophils.
- Author
-
Werner LM, Alcott A, Mohlin F, Ray JC, Belcher Dufrisne M, Smirnov A, Columbus L, Blom AM, and Criss AK
- Subjects
- Humans, Neutrophils microbiology, Complement C4b-Binding Protein metabolism, Bacterial Outer Membrane Proteins metabolism, Neisseria gonorrhoeae metabolism, Gonorrhea microbiology
- Abstract
Neisseria gonorrhoeae (Gc) is a human-specific pathogen that causes the sexually transmitted infection gonorrhea. Gc survives in neutrophil-rich gonorrheal secretions, and recovered bacteria predominantly express phase-variable, surface-expressed opacity-associated (Opa) proteins (Opa+). However, expression of Opa proteins like OpaD decreases Gc survival when exposed to human neutrophils ex vivo. Here, we made the unexpected observation that incubation with normal human serum, which is found in inflamed mucosal secretions, enhances survival of Opa+ Gc from primary human neutrophils. We directly linked this phenomenon to a novel complement-independent function for C4b-binding protein (C4BP). When bound to the bacteria, C4BP was necessary and sufficient to suppress Gc-induced neutrophil reactive oxygen species production and prevent neutrophil phagocytosis of Opa+ Gc. This research identifies for the first time a complement-independent role for C4BP in enhancing the survival of a pathogenic bacterium from phagocytes, thereby revealing how Gc exploits inflammatory conditions to persist at human mucosal surfaces., Competing Interests: The authors have no competing interests to declare., (Copyright: © 2023 Werner et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
39. Intracellular complement: Evidence, definitions, controversies, and solutions.
- Author
-
King BC and Blom AM
- Subjects
- Humans, Complement System Proteins metabolism
- Abstract
The term "intracellular complement" has been introduced recently as an umbrella term to distinguish functions of complement proteins that take place intracellularly, rather than in the extracellular environment. However, this rather undefined term leaves some confusion as to the classification of what intracellular complement really is, and as to which intracellular compartment(s) it should refer to. In this review, we will describe the evidence for both canonical and non-canonical functions of intracellular complement proteins, as well as the current controversies and unanswered questions as to the nature of the intracellular complement. We also suggest new terms to facilitate the accurate description and discussion of specific forms of intracellular complement and call for future experiments that will be required to provide more definitive evidence and a better understanding of the mechanisms of intracellular complement activity., (© 2022 The Authors. Immunological Reviews published by John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
40. Sushi domain-containing protein 4 binds to epithelial growth factor receptor and initiates autophagy in an EGFR phosphorylation independent manner.
- Author
-
Papadakos KS, Ekström A, Slipek P, Skourti E, Reid S, Pietras K, and Blom AM
- Subjects
- Humans, Animals, Mice, Phosphorylation, ErbB Receptors genetics, ErbB Receptors metabolism, Receptors, Growth Factor metabolism, Autophagy, Cell Line, Tumor, Triple Negative Breast Neoplasms metabolism
- Abstract
Background: Sushi domain-containing protein 4 (SUSD4) is a recently discovered protein with unknown cellular functions. We previously revealed that SUSD4 can act as complement inhibitor and as a potential tumor suppressor., Methods: In a syngeneic mouse model of breast cancer, tumors expressing SUSD4 had a smaller volume compared with the corresponding mock control tumors. Additionally, data from three different expression databases and online analysis tools confirm that for breast cancer patients, high mRNA expression of SUSD4 in the tumor tissue correlates with a better prognosis. In vitro experiments utilized triple-negative breast cancer cell lines (BT-20 and MDA-MB-468) stably expressing SUSD4. Moreover, we established a cell line based on BT-20 in which the gene for EGFR was knocked out with the CRISPR-Cas9 method., Results: We discovered that the Epithelial Growth Factor Receptor (EGFR) interacts with SUSD4. Furthermore, triple-negative breast cancer cell lines stably expressing SUSD4 had higher autophagic flux. The initiation of autophagy required the expression of EGFR but not phosphorylation of the receptor. Expression of SUSD4 in the breast cancer cells led to activation of the tumor suppressor LKB1 and consequently to the activation of AMPKα1. Finally, autophagy was initiated after stimulation of the ULK1, Atg14 and Beclin-1 axis in SUSD4 expressing cells., Conclusions: In this study we provide novel insight into the molecular mechanism of action whereby SUSD4 acts as an EGFR inhibitor without affecting the phosphorylation of the receptor and may potentially influence the recycling of EGFR to the plasma membrane., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
41. Expression of cartilage oligomeric matrix protein in periampullary adenocarcinoma is associated with pancreatobiliary-type morphology, higher levels of fibrosis and immune cell exclusion.
- Author
-
Papadakos KS, Lundgren S, Gialeli C, Micke P, Mezheyeuski A, Elebro J, Jirström K, and Blom AM
- Subjects
- Duodenal Neoplasms, Fibrosis genetics, Fibrosis immunology, Fibrosis pathology, Gene Expression, Humans, Pancreatic Neoplasms, Adenocarcinoma genetics, Adenocarcinoma immunology, Adenocarcinoma pathology, Ampulla of Vater immunology, Ampulla of Vater pathology, Cartilage Oligomeric Matrix Protein genetics, Cartilage Oligomeric Matrix Protein immunology, Common Bile Duct Neoplasms genetics, Common Bile Duct Neoplasms immunology, Common Bile Duct Neoplasms pathology
- Abstract
Cartilage oligomeric matrix protein (COMP) is an emerging regulator of tumor progression. The aim of this study was to evaluate the expression of COMP in periampullary adenocarcinoma with respect to prognostic value for survival and relapse, levels of fibrosis and infiltrating immune cells. COMP expression was evaluated using immunohistochemistry in primary tumors and subsets of paired lymph node metastases in tissue microarrays including 175 patients with periampullary adenocarcinoma. Collagen content was assessed with Sirius Red-Fast Green staining. High COMP levels were detected in cancer cells and in stroma, in 46% and 57% of the patients, respectively. High COMP expression was strongly associated with more aggressive pancreatobiliary-type (PB-type) compared to intestinal-type tumors (p < .0001). Importantly, high expression of COMP correlated with the exclusion of cytotoxic T-cells from the cancer cell compartment of the tumors, particularly in PB-type tumors. Higher levels of fibrosis measured by the density of collagen fibers correlated with high COMP levels in both cancer cells and stroma. This in turn could lead to exclusion of cytotoxic T-cells from accessing the cancer cells, a recognized immunotherapy resistance mechanism. Targeting COMP could therefore be considered as a novel therapeutic strategy in PB-type periampullary adenocarcinoma., Competing Interests: No potential conflict of interest was reported by the author(s)., (© 2022 The Author(s). Published with license by Taylor & Francis Group, LLC.)
- Published
- 2022
- Full Text
- View/download PDF
42. Complement C4 Copy Number Variation is Linked to SSA/Ro and SSB/La Autoantibodies in Systemic Inflammatory Autoimmune Diseases.
- Author
-
Lundtoft C, Pucholt P, Martin M, Bianchi M, Lundström E, Eloranta ML, Sandling JK, Sjöwall C, Jönsen A, Gunnarsson I, Rantapää-Dahlqvist S, Bengtsson AA, Leonard D, Baecklund E, Jonsson R, Hammenfors D, Forsblad-d'Elia H, Eriksson P, Mandl T, Magnusson Bucher S, Norheim KB, Auglaend Johnsen SJ, Omdal R, Kvarnström M, Wahren-Herlenius M, Notarnicola A, Andersson H, Molberg Ø, Diederichsen LP, Almlöf J, Syvänen AC, Kozyrev SV, Lindblad-Toh K, Nilsson B, Blom AM, Lundberg IE, Nordmark G, Diaz-Gallo LM, Svenungsson E, and Rönnblom L
- Subjects
- Autoantibodies genetics, Complement C4 genetics, Complement C4b genetics, DNA Copy Number Variations, Humans, Risk Factors, Lupus Erythematosus, Systemic genetics, Myositis
- Abstract
Objective: Copy number variation of the C4 complement components, C4A and C4B, has been associated with systemic inflammatory autoimmune diseases. This study was undertaken to investigate whether C4 copy number variation is connected to the autoimmune repertoire in systemic lupus erythematosus (SLE), primary Sjögren's syndrome (SS), or myositis., Methods: Using targeted DNA sequencing, we determined the copy number and genetic variants of C4 in 2,290 well-characterized Scandinavian patients with SLE, primary SS, or myositis and 1,251 healthy controls., Results: A prominent relationship was observed between C4A copy number and the presence of SSA/SSB autoantibodies, which was shared between the 3 diseases. The strongest association was detected in patients with autoantibodies against both SSA and SSB and 0 C4A copies when compared to healthy controls (odds ratio [OR] 18.0 [95% confidence interval (95% CI) 10.2-33.3]), whereas a weaker association was seen in patients without SSA/SSB autoantibodies (OR 3.1 [95% CI 1.7-5.5]). The copy number of C4 correlated positively with C4 plasma levels. Further, a common loss-of-function variant in C4A leading to reduced plasma C4 was more prevalent in SLE patients with a low copy number of C4A. Functionally, we showed that absence of C4A reduced the individuals' capacity to deposit C4b on immune complexes., Conclusion: We show that a low C4A copy number is more strongly associated with the autoantibody repertoire than with the clinically defined disease entities. These findings may have implications for understanding the etiopathogenetic mechanisms of systemic inflammatory autoimmune diseases and for patient stratification when taking the genetic profile into account., (© 2022 The Authors. Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)
- Published
- 2022
- Full Text
- View/download PDF
43. Alternative splicing encodes functional intracellular CD59 isoforms that mediate insulin secretion and are down-regulated in diabetic islets.
- Author
-
Golec E, Ekström A, Noga M, Omar-Hmeadi M, Lund PE, Villoutreix BO, Krus U, Wozniak K, Korsgren O, Renström E, Barg S, King BC, and Blom AM
- Subjects
- CD59 Antigens genetics, CD59 Antigens metabolism, Humans, Insulin Secretion, Protein Isoforms genetics, Protein Isoforms metabolism, Alternative Splicing, Diabetes Mellitus genetics
- Abstract
Human pancreatic islets highly express CD59, which is a glycosylphosphatidylinositol (GPI)-anchored cell-surface protein and is required for insulin secretion. How cell-surface CD59 could interact with intracellular exocytotic machinery has so far not been described. We now demonstrate the existence of CD59 splice variants in human pancreatic islets, which have unique C-terminal domains replacing the GPI-anchoring signal sequence. These isoforms are found in the cytosol of β-cells, interact with SNARE proteins VAMP2 and SNAP25, colocalize with insulin granules, and rescue insulin secretion in CD59-knockout (KO) cells. We therefore named these isoforms IRIS-1 and IRIS-2 (Isoforms Rescuing Insulin Secretion 1 and 2). Antibodies raised against each isoform revealed that expression of both IRIS-1 and IRIS-2 is significantly lower in islets isolated from human type 2 diabetes (T2D) patients, as compared to healthy controls. Further, glucotoxicity induced in primary, healthy human islets led to a significant decrease of IRIS-1 expression, suggesting that hyperglycemia (raised glucose levels) and subsequent decreased IRIS-1 expression may contribute to relative insulin deficiency in T2D patients. Similar isoforms were also identified in the mouse CD59B gene, and targeted CRISPR/Cas9-mediated knockout showed that these intracellular isoforms, but not canonical CD59B, are involved in insulin secretion from mouse β-cells. Mouse IRIS-2 is also down-regulated in diabetic db/db mouse islets. These findings establish the endogenous existence of previously undescribed non–GPI-anchored intracellular isoforms of human CD59 and mouse CD59B, which are required for normal insulin secretion.
- Published
- 2022
- Full Text
- View/download PDF
44. Alternative translation and retrotranslocation of cytosolic C3 that detects cytoinvasive bacteria.
- Author
-
Kremlitzka M, Colineau L, Nowacka AA, Mohlin FC, Wozniak K, Blom AM, and King BC
- Subjects
- Bacteria metabolism, Cytosol metabolism, Endoplasmic Reticulum metabolism, Complement C3 metabolism, Proteasome Endopeptidase Complex metabolism
- Abstract
Complement C3 was originally regarded as a serum effector protein, although recent data has emerged suggesting that intracellular C3 can also regulate basic cellular processes. Despite the growing interest in intracellular C3 functions, the mechanism behind its generation has not been demonstrated. In this study we show that C3 can be expressed from an alternative translational start site, resulting in C3 lacking the signal peptide, which is therefore translated in the cytosol. In contrast to the secreted form, alternatively translated cytosolic C3 is not glycosylated, is present mainly in a reduced state, and is turned over by the ubiquitin-proteasome system. C3 can also be retrotranslocated from the endoplasmic reticulum into the cytosol, structurally resembling secreted C3. Finally, we demonstrate that intracellular cytosolic C3 can opsonize invasive Staphylococcus aureus within epithelial cell, slowing vacuolar escape as well as impacting bacterial survival on subsequent exposure to phagocytes. Our work therefore reveals the existence and origin of intracellular, cytosolic C3, and demonstrates functions for cytosolic C3 in intracellular detection of cytoinvasive pathogens., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
45. The Hidden Side of Complement Regulator C4BP: Dissection and Evaluation of Its Immunomodulatory Activity.
- Author
-
Serrano I, Luque A, Mitjavila F, Blom AM, Rodríguez de Córdoba S, Vega MC, Torras J, and Aran JM
- Subjects
- Cytokines, Humans, Immunomodulation, Monocytes metabolism, Complement C4b-Binding Protein metabolism, Lupus Nephritis
- Abstract
C4b-binding protein (C4BP) is a well-known regulator of the complement system that holds additional and important activities unrelated to complement inhibition. Recently, we have described a novel immunomodulatory activity in the minor C4BP(β-) isoform directly acting over inflammatory phagocytes. Here we show that incorporation of the β-chain to the C4BP α-chain oligomer interferes with this immunomodulatory activity of C4BP. Moreover, an oligomeric form including only the complement control protein 6 (CCP6) domain of the C4BP α-chain (PRP6-HO7) is sufficient to "reprogram" monocyte-derived DCs (Mo-DCs) from a pro-inflammatory and immunogenic phenotype to an anti-inflammatory and tolerogenic state. PRP6-HO7 lacks complement regulatory activity but retains full immunomodulatory activity over inflammatory Mo-DCs induced by TLRs, characterized by downregulation of relevant surface markers such as CD83, HLA-DR, co-stimulatory molecules such as CD86, CD80 and CD40, and pro-inflammatory cytokines such as IL-12 and TNF-α. Furthermore, PRP6-HO7-treated Mo-DCs shows increased endocytosis, significantly reduced CCR7 expression and CCL21-mediated chemotaxis, and prevents T cell alloproliferation. Finally, PRP6-HO7 shows also full immunomodulatory activity over Mo-DCs isolated from lupus nephritis patients with active disease, even without further pro-inflammatory stimulation. Therefore PRP6-HO7, retaining the immunomodulatory activity of C4BP(β-) and lacking its complement regulatory activity, might represent a promising and novel alternative to treat autoimmune diseases., Competing Interests: IS, AL and JA are co-inventors on pending or issued patents involving compounds and methods for immunomodulation. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Serrano, Luque, Mitjavila, Blom, Rodríguez de Córdoba, Vega, Torras and Aran.)
- Published
- 2022
- Full Text
- View/download PDF
46. Biomarkers of Complement and Platelet Activation are not correlated with the One or Twenty-Four Hours Corrected Count Increments in Prophylactically Platelet Transfused Hematological Patients: a Prospective Cohort Study.
- Author
-
Åkesson A, Ljungkvist M, Martin M, Blom AM, Klintman J, Schött U, Zetterberg E, and Kander T
- Subjects
- Adult, Aged, Cohort Studies, Female, Humans, Male, Middle Aged, Prospective Studies, Time Factors, Biomarkers metabolism, Complement System Proteins physiology, Platelet Activation physiology, Platelet Transfusion methods
- Abstract
Platelet transfusion refractoriness is a serious clinical concern that complicates the management of thrombocytopenic patients. Previous studies have suggested a potential role for both complement and platelet activation based on in vitro analyses of platelet concentrates. In this study, the post-transfusion platelet response, as indicated by the corrected count increment at 1 and 24 h after prophylactic platelet transfusions, respectively, was correlated with the 1 h post-transfusion Δconcentration (1 h post-transfusion - pretransfusion) of complement and platelet activation biomarkers. The study was registered as a clinical trial at ClinicalTrials.gov (identifier: NCT02601131) and patients were recruited during inpatient care in the hematological department. Soluble terminal complement complexes, soluble P-selectin and soluble CD40 ligand were analyzed. Confirmed alloimmunized patients were excluded. Included subjects were either given platelet transfusions (n = 43) and categorized into four clinical study groups or included in a non-transfused control group (n = 10). In total, 54 transfusions were included. No transfusion-mediated complement activation was observed. The transfusions were associated with a significant increase in the concentration of soluble P-selectin ( p < .001), primarily corresponding to the passive infusion of soluble P-selectin-containing plasma residuals. The Δconcentration of soluble P-selectin was, however, not significantly correlated with the corrected count increments. Thus, significant correlations between biomarkers of complement and platelet activation and the post-transfusion platelet response could not be demonstrated in this study.
- Published
- 2022
- Full Text
- View/download PDF
47. Complement Factor H-Related Proteins FHR1 and FHR5 Interact With Extracellular Matrix Ligands, Reduce Factor H Regulatory Activity and Enhance Complement Activation.
- Author
-
Papp A, Papp K, Uzonyi B, Cserhalmi M, Csincsi ÁI, Szabó Z, Bánlaki Z, Ermert D, Prohászka Z, Erdei A, Ferreira VP, Blom AM, and Józsi M
- Subjects
- Extracellular Matrix, Humans, Inflammation, Ligands, Complement Activation, Complement C3b Inactivator Proteins metabolism, Complement Factor H, Complement System Proteins metabolism
- Abstract
Components of the extracellular matrix (ECM), when exposed to body fluids may promote local complement activation and inflammation. Pathologic complement activation at the glomerular basement membrane and at the Bruch's membrane is implicated in renal and eye diseases, respectively. Binding of soluble complement inhibitors to the ECM, including factor H (FH), is important to prevent excessive complement activation. Since the FH-related (FHR) proteins FHR1 and FHR5 are also implicated in these diseases, our aim was to study whether these FHRs can also bind to ECM components and affect local FH activity and complement activation. Both FH and the FHRs showed variable binding to ECM components. We identified laminin, fibromodulin, osteoadherin and PRELP as ligands of FHR1 and FHR5, and found that FHR1 bound to these ECM components through its C-terminal complement control protein (CCP) domains 4-5, whereas FHR5 bound via its middle region, CCPs 3-7. Aggrecan, biglycan and decorin did not bind FH, FHR1 and FHR5. FHR5 also bound to immobilized C3b, a model of surface-deposited C3b, via CCPs 3-7. By contrast, soluble C3, C3(H
2 O), and the C3 fragments C3b, iC3b and C3d bound to CCPs 8-9 of FHR5. Properdin, which was previously described to bind via CCPs 1-2 to FHR5, did not bind in its physiologically occurring serum forms in our assays. FHR1 and FHR5 inhibited the binding of FH to the identified ECM proteins in a dose-dependent manner, which resulted in reduced FH cofactor activity. Moreover, both FHR1 and FHR5 enhanced alternative complement pathway activation on immobilized ECM proteins when exposed to human serum, resulting in the increased deposition of C3-fragments, factor B and C5b-9. Thus, our results identify novel ECM ligands of FH family proteins and indicate that FHR1 and FHR5 are competitive inhibitors of FH on ECM and, when bound to these ligands, they may enhance local complement activation and promote inflammation under pathological conditions., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Papp, Papp, Uzonyi, Cserhalmi, Csincsi, Szabó, Bánlaki, Ermert, Prohászka, Erdei, Ferreira, Blom and Józsi.)- Published
- 2022
- Full Text
- View/download PDF
48. Cellular and immunological mechanisms influence host-adapted phenotypes in a vector-borne microparasite.
- Author
-
Lin YP, Tufts DM, Combs M, Dupuis AP 2nd, Marcinkiewicz AL, Hirsbrunner AD, Diaz AJ, Stout JL, Blom AM, Strle K, Davis AD, Kramer LD, Kolokotronis SO, and Diuk-Wasser MA
- Subjects
- Animals, Host Adaptation, Peromyscus, Phenotype, Borrelia burgdorferi genetics, Borrelia burgdorferi Group genetics, Lyme Disease
- Abstract
Predicting pathogen emergence and spillover risk requires understanding the determinants of a pathogens' host range and the traits involved in host competence. While host competence is often considered a fixed species-specific trait, it may be variable if pathogens diversify across hosts. Balancing selection can lead to maintenance of pathogen polymorphisms (multiple-niche-polymorphism; MNP). The causative agent of Lyme disease, Borrelia burgdorferi ( Bb ), provides a model to study the evolution of host adaptation, as some Bb strains defined by their outer surface protein C ( ospC ) genotype, are widespread in white-footed mice and others are associated with non-rodent vertebrates (e.g. birds). To identify the mechanisms underlying potential strain × host adaptation, we infected American robins and white-footed mice, with three Bb strains of different ospC genotypes. Bb burdens varied by strain in a host-dependent fashion, and strain persistence in hosts largely corresponded to Bb survival at early infection stages and with transmission to larvae (i.e. fitness). Early survival phenotypes are associated with cell adhesion, complement evasion and/or inflammatory and antibody-mediated removal of Bb, suggesting directional selective pressure for host adaptation and the potential role of MNP in maintaining OspC diversity. Our findings will guide future investigations to inform eco-evolutionary models of host adaptation for microparasites.
- Published
- 2022
- Full Text
- View/download PDF
49. Functional Analysis of Variants in Complement Factor I Identified in Age-Related Macular Degeneration and Atypical Hemolytic Uremic Syndrome.
- Author
-
de Jong S, de Breuk A, Bakker B, Katti S, Hoyng CB, Nilsson SC, Blom AM, van den Heuvel LP, den Hollander AI, and Volokhina EB
- Subjects
- Amino Acid Substitution, Complement C3b immunology, Complement C3b metabolism, Female, Humans, Male, Atypical Hemolytic Uremic Syndrome blood, Atypical Hemolytic Uremic Syndrome genetics, Atypical Hemolytic Uremic Syndrome immunology, Complement Factor I genetics, Complement Factor I immunology, Complement Factor I metabolism, Macular Degeneration blood, Macular Degeneration genetics, Macular Degeneration immunology, Mutation, Missense
- Abstract
Complement factor I (FI) is a central inhibitor of the complement system, and impaired FI function increases complement activation, contributing to diseases such as age-related macular degeneration (AMD) and atypical hemolytic uremic syndrome (aHUS). Genetic variation in complement factor I ( CFI ) has been identified in both AMD and aHUS, with more than half of these variants leading to reduced FI secretion levels. For many of the variants with normal FI secretion, however, functional implications are not yet known. Here we studied 11 rare missense variants, with FI secretion levels comparable to wildtype, but a predicted damaging effects based on the Combined Annotation Dependent Depletion (CADD) score. Three variants (p.Pro50Ala, p.Arg339Gln, and p.Ser570Thr) were analyzed in plasma and serum samples of carriers affected by AMD. All 11 variants (nine for the first time in this study) were recombinantly expressed and the ability to degrade C3b was studied with the C3b degradation assay. The amount of degradation was determined by measuring the degradation product iC3b with ELISA. Eight of 11 (73%) mutant proteins (p.Pro50Ala, p.Arg339Gln, p.Ile340Thr, p.Gly342Glu, p.Gly349Arg, p.Arg474Gln, p.Gly487Cys, and p.Gly512Ser) showed significantly impaired C3b degradation, and were therefore classified as likely pathogenic. Our data indicate that genetic variants in CFI with a CADD score >20 are likely to affect FI function, and that monitoring iC3b in a degradation assay is a useful tool to establish the pathogenicity of CFI variants in functional studies., Competing Interests: Author SK was employed by company Gemini Therapeutics Inc.AH is a consultant for Ionis Pharmaceuticals, Gyroscope Therapeutics, Gemini Therapeutics and F. Hoffmann-La Roche. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. This study received funding from Gemini Therapeutics Inc. The funder’s employer (SK) had contributed to study design and preparation of the manuscript., (Copyright © 2022 de Jong, de Breuk, Bakker, Katti, Hoyng, Nilsson, Blom, van den Heuvel, den Hollander and Volokhina.)
- Published
- 2022
- Full Text
- View/download PDF
50. Clinical characterization and identification of rare genetic variants in atypical hemolytic uremic syndrome: A Swedish retrospective observational study.
- Author
-
Åkesson A, Martin M, Blom AM, Rossing M, Gabrielaite M, Zetterberg E, and Klintman J
- Subjects
- Adult, Female, Humans, Male, Middle Aged, Retrospective Studies, Sweden, Young Adult, Atypical Hemolytic Uremic Syndrome genetics, Genetic Variation genetics
- Abstract
Complement-mediated atypical hemolytic uremic syndrome (aHUS) is an ultra-rare renal disease primarily caused by genetic alterations in complement proteins. The genetic work-up required for confirmation of diagnosis is complicated and not always logistically accessible. The aim of the present study was to apply a diagnostic scheme compliant with the American College of Medical Genetics and Genomics guidelines to investigate the prevalence of complement-mediated aHUS among subjects formerly included in a retrospective cohort of clinically suspected aHUS. Clinical outcomes and genetic correlations to complement analyses were assessed. Subjects were investigated with medical record reviewing, inquiries, and laboratory analyses composed of whole genome sequencing; enzyme-linked immunosorbent assays for factor I, factor H, and factor H-specific antibodies; nephelometry for complement components three of four; flow cytometry for CD46 surface expression and immunoblotting for the presence of factor H-related protein 1. In total, 45% (n = 60/134) of the subjects were deceased at the time of study. Twenty of the eligible subjects consented to study participation. Based on genetic sequencing and clinical characteristics, six were categorized as definite/highly suspected complement-mediated aHUS, 10 as non-complement-mediated aHUS and four as having an HUS-like phenotype. In the complement-mediated aHUS group, two subjects had not received an aHUS diagnosis during the routine clinical management. Disease-contributing/likely disease-contributing genetic variants were identified in five subjects, including a novel missense variant in the complement factor H gene (c.3450A>G, p.I1150M). This study illustrates the risk for misdiagnosis in the management of patients with complement-mediated aHUS and the importance of a comprehensive assessment of both phenotype and genotype to reach a diagnosis., (© 2021 The Authors. Therapeutic Apheresis and Dialysis published by John Wiley & Sons Australia, Ltd on behalf of International Society for Apheresis, Japanese Society for Apheresis, and Japanese Society for Dialysis Therapy.)
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.