Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Stem Cell Res Ther ; 15(1): 135, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715130

ABSTRACT

BACKGROUND: Biomaterials used in bone tissue engineering must fulfill the requirements of osteoconduction, osteoinduction, and osseointegration. However, biomaterials with good osteoconductive properties face several challenges, including inadequate vascularization, limited osteoinduction and barrier ability, as well as the potential to trigger immune and inflammatory responses. Therefore, there is an urgent need to develop guided bone regeneration membranes as a crucial component of tissue engineering strategies for repairing bone defects. METHODS: The mZIF-8/PLA membrane was prepared using electrospinning technology and simulated body fluid external mineralization method. Its ability to induce biomimetic mineralization was evaluated through TEM, EDS, XRD, FT-IR, zeta potential, and wettability techniques. The biocompatibility, osteoinduction properties, and osteo-immunomodulatory effects of the mZIF-8/PLA membrane were comprehensively evaluated by examining cell behaviors of surface-seeded BMSCs and macrophages, as well as the regulation of cellular genes and protein levels using PCR and WB. In vivo, the mZIF-8/PLA membrane's potential to promote bone regeneration and angiogenesis was assessed through Micro-CT and immunohistochemical staining. RESULTS: The mineralized deposition enhances hydrophilicity and cell compatibility of mZIF-8/PLA membrane. mZIF-8/PLA membrane promotes up-regulation of osteogenesis and angiogenesis related factors in BMSCs. Moreover, it induces the polarization of macrophages towards the M2 phenotype and modulates the local immune microenvironment. After 4-weeks of implantation, the mZIF-8/PLA membrane successfully bridges critical bone defects and almost completely repairs the defect area after 12-weeks, while significantly improving the strength and vascularization of new bone. CONCLUSIONS: The mZIF-8/PLA membrane with dual osteoconductive and immunomodulatory abilities could pave new research paths for bone tissue engineering.


Subject(s)
Bone Regeneration , Bone Regeneration/drug effects , Animals , Osteogenesis/drug effects , Tissue Engineering/methods , Biocompatible Materials/pharmacology , Biocompatible Materials/chemistry , Mice , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Membranes, Artificial , Guided Tissue Regeneration/methods , Tissue Scaffolds/chemistry , Polyesters/chemistry , Polyesters/pharmacology , Rats
2.
Front Immunol ; 15: 1368322, 2024.
Article in English | MEDLINE | ID: mdl-38558821

ABSTRACT

Introduction: Activation of complement through the alternative pathway (AP) has a key role in the pathogenesis of IgA nephropathy (IgAN). We previously showed, by intraperitoneal injection of Lactobacillus casei cell wall extract (LCWE), C57BL/6 mice develop mild kidney damage in association with glomerular IgA deposition. To further address complement activity in causing glomerular histological alterations as suggested in the pathogenesis of IgAN, here we used mice with factor H mutation (FHW/R) to render AP overactivation in conjunction with LCWE injection to stimulate intestinal production of IgA. Methods: Dose response to LCWE were examined between two groups of FHW/R mice. Wild type (FHW/W) mice stimulated with LCWE were used as model control. Results: The FHW/R mice primed with high dose LCWE showed elevated IgA and IgA-IgG complex levels in serum. In addition to 100% positive rate of IgA and C3, they display elevated biomarkers of kidney dysfunction, coincided with severe pathological lesions, resembling those of IgAN. As compared to wild type controls stimulated by the same high dose LCWE, these FHW/R mice exhibited stronger complement activation in the kidney and in circulation. Discussion: The new mouse model shares many disease features with IgAN. The severity of glomerular lesions and the decline of kidney functions are further aggravated through complement overactivation. The model may be a useful tool for preclinical evaluation of treatment response to complement-inhibitors.


Subject(s)
Glomerulonephritis, IGA , Lacticaseibacillus casei , Mice , Animals , Complement Factor H/genetics , Mice, Inbred C57BL , Glomerulonephritis, IGA/pathology , Complement System Proteins/genetics , Immunoglobulin A , Mutation
3.
Kidney Blood Press Res ; 49(1): 20-26, 2024.
Article in English | MEDLINE | ID: mdl-38048756

ABSTRACT

INTRODUCTION: High serum phosphorus level has been reported to be a risk factor for disease progression in patients with chronic kidney disease, whereas, its role in IgA nephropathy (IgAN) still remains uncertain. This study aimed to investigate the association between serum phosphorus and progression of IgAN. METHODS: A total of 247 patients diagnosed with IgAN from 2016.11 to 2019.12 at the First Affiliated Hospital of Xi'an Jiaotong University were retrospectively enrolled in this study. The association between serum phosphorus and kidney disease progression events, defined as 30% estimated glomerular filtration rate (eGFR) decline or kidney failure, was evaluated using Cox models. RESULTS: Serum phosphorus was an independent risk factor for poor renal outcome after adjusting for age, gender, urine protein, MAP, eGFR, hemoglobin, Oxford S and T scores (HR, 2.586; 95% CI, 1.238-5.400, p = 0.011). The addition of serum phosphorus to the reference model containing clinical and pathological variables significantly improved the risk prediction of IgAN progression (C statistic, 0.836; 95% CI, 0.783-0.889) as compared with the reference model (C statistic, 0.821; 95% CI, 0.756-0.886). The ability of serum phosphorus level to predict progression was much stronger in IgAN patients without use of immunosuppression (HR 5.173; 95% CI, 1.791-14.944; p = 0.002). CONCLUSION: Higher serum phosphorus levels were independently associated with kidney disease progression in patients with IgAN, especially in those without immunosuppression. The addition of serum phosphorus to clinical and pathological data at the time of biopsy significantly improved risk prediction of IgAN progression.


Subject(s)
Glomerulonephritis, IGA , Kidney Failure, Chronic , Humans , Glomerulonephritis, IGA/diagnosis , Retrospective Studies , Follow-Up Studies , Disease Progression , Kidney/pathology , Glomerular Filtration Rate , Kidney Failure, Chronic/complications , Prognosis
6.
Clin Exp Nephrol ; 27(12): 1060-1066, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37668806

ABSTRACT

BACKGROUND: Phospholipase A2 receptor (PLA2R)-associated membranous nephropathy (MN) was manifested as seropositive for PLA2R antibodies (SAb) and/or glomerular PLA2R antigens' (GAg) deposits. According to the test of SAb and GAg, PLA2R-associated MN can be divided into SAb + /GAg-, SAb-/GAg + , and SAb + /GAg + groups. The clinical characteristics and outcomes of the three groups need to be further evaluated. METHODS: 184 PLA2R-associated MN patients were enrolled. SAb was measured by enzyme-linked immunosorbent assay with a cut-off value of 14 RU/mL. GAg was detected by immunofluorescence using a paraffin section of renal biopsy samples. Clinical characteristics and the decline of eGFR were compared among the 3 groups. RESULTS: There were 33 SAb + /GAg-, 46 SAb-/GAg +, and 105 SAb + /GAg + PLA2R-associated MN patients reviewed. Clinical characteristics, such as the level of proteinuria, serum albumin, as well as eGFR, were comparable between the SAb + /GAg- and SAb + /GAg + patients. While SAb-/GAg + patients exhibited mild clinical manifestations as evidenced by higher serum albumin (P < 0.001) and lower proteinuria (p = 0.049) compared with SAb + /GAg + patients. After 21.96 ± 7.39 month follow-up, the eGFR decrease was no difference between the SAb + /GAg- and SAb + /GAg + patients. SAb-/GAg + patients had a lower rate of the > 20% eGFR decline as well as a 50% eGFR decline compared with the SAb + /GAg + patients (10.87% vs 30.48%, p = 0.013; 0.00% vs 4.76%, p = 0.324). CONCLUSIONS: Our study showed that the clinical manifestations of SAb + /Gag- patients were the same as those of double-positive patients, while SAb-/GAg + patients exhibited mild clinical manifestations and slower eGFR decline compared to the double-positive patients.


Subject(s)
Glomerulonephritis, Membranous , Humans , Receptors, Phospholipase A2 , Proteinuria/etiology , Autoantibodies , Serum Albumin
7.
Ren Fail ; 45(1): 2235431, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37470370

ABSTRACT

Purpose: To investigate the clinical characteristics, pathological features, and outcomes of patients with antineutrophil cytoplasmic antibody (ANCA)-positive systemic lupus erythematosus (SLE) in northwest China.Methods: This retrospective study included 491 patients with SLE tested for ANCA antibodies and 171 patients with ANCA-associated vasculitis (AAV) as controls. Subgroup analysis limited to those with renal involvement, and by ANCA antibody subtype (PR3 vs MPO). To compare the proteinuria remission rates between ANCA-positive and ANCA-negative lupus nephritis (LN) groups, a logistic regression model was used for propensity score matching based on age, hemoglobin, and baseline estimated glomerular filtration rate (eGFR).Results: Compared to ANCA-negative SLE (n = 442), ANCA-positive SLE (n = 46) occur in older patients; however, these patients were younger than those with AAV (n = 167). The eGFR of patients with ANCA-positive LN (n = 25) was higher than that of patients having AAV with renal involvement (n = 56) but lower than that of patients with ANCA-negative LN (n = 163). Patients with SLE who had MPO-ANCA (n = 16) had higher levels of serum creatinine compared to those with PR3-ANCA (n = 30) (156.5 µmol/L vs. 45.5 µmol/L, p = 0.005). During the follow-up period, the remission rate of proteinuria in patients with ANCA-positive LN was lower than that of patients with ANCA-negative LN (50% vs. 75%, p = 0.008).Conclusion: Patients with ANCA-positive LN may have worse baseline renal function and lower protein remission rates compared to patients with ANCA-negative LN. ANCA titers should be regularly monitored throughout the follow-up period in patients with SLE, especially in cases of renal involvement.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis , Lupus Erythematosus, Systemic , Lupus Nephritis , Humans , Aged , Antibodies, Antineutrophil Cytoplasmic , Retrospective Studies , Lupus Erythematosus, Systemic/complications , Lupus Nephritis/complications , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/complications , Proteinuria/complications , Peroxidase
9.
Front Immunol ; 13: 1013990, 2022.
Article in English | MEDLINE | ID: mdl-36189210

ABSTRACT

Monoclonal immunoglobulin produced by clonal plasma cells is the main cause in multiple myeloma and monoclonal gammopathy of renal significance. Because of the complicated purification method and the low stoichiometry of purified protein and glycans, site-specific N-glycosylation characterization for monoclonal immunoglobulin is still challenging. To profile the site-specific N-glycosylation of monoclonal immunoglobulins is of great interest. Therefore, in this study, we presented an integrated workflow for micro monoclonal IgA and IgG purification from patients with multiple myeloma in the HYDRASYS system, in-agarose-gel digestion, LC-MS/MS analysis without intact N-glycopeptide enrichment, and compared the identification performance of different mass spectrometry dissociation methods (EThcD-sceHCD, sceHCD, EThcD and sceHCD-pd-ETD). The results showed that EThcD-sceHCD was a better choice for site-specific N-glycosylation characterization of micro in-agarose-gel immunoglobulins (~2 µg) because it can cover more unique intact N-glycopeptides (37 and 50 intact N-glycopeptides from IgA1 and IgG2, respectively) and provide more high-quality spectra than sceHCD, EThcD and sceHCD-pd-ETD. We demonstrated the benefits of the alternative strategy in site-specific N-glycosylation characterizing micro monoclonal immunoglobulins obtained from bands separated by electrophoresis. This work could promote the development of clinical N-glycoproteomics and related immunology.


Subject(s)
Multiple Myeloma , Tandem Mass Spectrometry , Chromatography, Liquid/methods , Glycopeptides , Glycosylation , Humans , Immunoglobulin A , Immunoglobulin G , Polysaccharides , Sepharose , Tandem Mass Spectrometry/methods
10.
Adv Healthc Mater ; 11(22): e2201877, 2022 11.
Article in English | MEDLINE | ID: mdl-36085440

ABSTRACT

Bioprinting specific tissues with robust viability is a great challenge, requiring a delicate balance between a densely cellular distribution and hydrogel network crosslinking density. Microtissues composed of tissue-specific mesenchymal stem cells and extra cellular matrix (ECM) particles provide an alternative scheme for realizing biomimetic cell density and microenvironment. Nevertheless, due to their instability during manufacturing, scarce efforts have been made to date to assemble them using rapid prototyping methods. Here, a novel microtissue bioink with good printability and cellular viability maintenance for digital light processing (DLP) bioprinting is introduced. Generally, the microtissue bioink is prepared by crosslinking acellular matrix microparticles and GelMA hydrogel with a specific proportion. The microtissue bioink exhibits the desired mechanical properties, swelling ratio, and has almost no influences on printability. For instance, a DLP bioprinted ear with a precise auricle structure using microtia chondrocytes microtissue boink is created. Additionally, the chondrocytes in the printed ears show obvious advantages in cell proliferation in vitro and auricular cartilage regeneration in vivo. The microtissue composite bioink for DLP printing not only enables accurate assembly of organ building blocks but also provides a 3D shelter to ensure printed cells' viability.


Subject(s)
Bioprinting , Chondrocytes , Tissue Engineering , Tissue Scaffolds/chemistry , Printing, Three-Dimensional , Hydrogels/chemistry
11.
Bio Protoc ; 12(13)2022 Jul 05.
Article in English | MEDLINE | ID: mdl-35937933

ABSTRACT

Both IgA nephropathy and IgA vasculitis, formerly known as Henoch-SchÓ§nlein purpura, are immune deposition diseases. IgA nephropathy is caused by the deposition of aberrantly formed poly-IgA complexes from blood circulation to the kidney glomerulus; IgA vasculitis is characterized by IgA-dominant immune deposits to small vessels of the skin and other organs, including the kidney. Therefore, measuring the disease-causing poly-IgA contents in the plasma is needed to study these conditions. However, while clinical tests for the level of total plasma IgA are routinely performed, methods for specific detection of poly-IgA contents are unavailable in clinical medicine. In this protocol, we describe a practical solution for measuring poly-IgA in patient samples. The new method is based on the biological selectivity of IgA Fcα receptor I (FcαRI/CD89) toward poly-IgA species, in contrast to its relatively low affinity for normal monomeric IgA. By devising recombinant CD89 ectodomain as the "capturing" probe, we validated the feasibility of the assay for measuring plasma poly-IgA levels in a 96-well format. The methodology was able to differentiate plasma samples of IgA nephropathy, or related IgA vasculitis, from those of other autoimmune kidney disease types or from healthy controls. Moreover, the measured poly-IgA indices not only correlated with the severity of IgA nephropathy, but the levels also trended lower following corticosteroid or immunosuppressant treatments of patients. Therefore, we anticipate the new assay will provide useful measurements of the IgA nephropathy disease activity index for stratifying disease severity or for evaluating treatment response. Graphical abstract.

12.
JCI Insight ; 7(7)2022 04 08.
Article in English | MEDLINE | ID: mdl-35393947

ABSTRACT

Amyloidosis involves stepwise growth of fibrils assembled from soluble precursors. Transthyretin (TTR) naturally folds into a stable tetramer, whereas conditions and mutations that foster aberrant monomer formations facilitate TTR oligomeric aggregation and subsequent fibril extension. We investigated the early assembly of oligomers by WT TTR compared with its V30M and V122I variants. We monitored time-dependent redistribution among monomer, dimer, tetramer, and oligomer contents in the presence and absence of multimeric TTR seeds. The seeds were artificially constructed recombinant multimers that contained 20-40 TTR subunits via engineered biotin-streptavidin (SA) interactions. As expected, these multimer seeds rapidly nucleated TTR monomers into larger complexes, while having less effect on dimers and tetramers. In vivo, SA-induced multimers formed TTR-like deposits in the heart and the kidney following i.v. injection in mice. While all 3 variants prominently deposited glomerulus in the kidney, only V30M resulted in extensive deposition in the heart. The cardiac TTR deposits varied in size and shape and were localized in the intermyofibrillar space along the capillaries. These results are consistent with the notion of monomeric TTR engaging in high-avidity interactions with tissue amyloids. Our multimeric induction approach provides a model for studying the initiation of TTR deposition in the heart.


Subject(s)
Amyloid , Prealbumin , Amyloid/chemistry , Amyloid/genetics , Amyloid/metabolism , Animals , Mice , Prealbumin/genetics
13.
J Am Soc Nephrol ; 33(5): 918-935, 2022 05.
Article in English | MEDLINE | ID: mdl-35172987

ABSTRACT

BACKGROUND: IgA nephropathy is a common primary glomerulonephritis caused by mesangial deposition of poly-IgA complexes. The disease follows a variable course of clinical progression, with a high risk of kidney failure. Although no specific therapy is available, enzymatic strategies to clear IgA deposits are being considered for the treatment of rapidly progressive IgA nephropathy. METHODS: We chose an IgA protease of commensal bacterium Clostridium ramosum, termed AK183, as the template for constructing a recombinant biologic. To extend the t1/2 in blood, we fused AK183 to the Fc segment of human IgG1. Activities of this Fc-AK183 fusion protein toward the cleavage and subsequent clearance of IgA were tested in mouse models. RESULTS: First, we discovered an autocleavage activity of AK183 that separates the N-terminal protease from its C-terminal autotransporter ß domain. Therefore, we grafted Fc to the N terminus of AK183 and demonstrated its week-long enzymatic activity in mice. In addition, the proteolytic fragments of IgA generated in the reaction with Fc-AK183 were effectively removed from circulation via kidney filtration. The combined actions of Fc-AK183-mediated cleavage and subsequent renal clearance of IgA resulted in a lasting obliteration of blood IgA, as demonstrated in a human IgA-injection model and in a humanized α1KI transgenic model. Fc-AK183 was also able to remove chronic IgA and associated complement C3 deposits in the glomerulus. CONCLUSION: We constructed a chimeric fusion of IgA protease with Fc and demonstrated its long-lasting efficacy as a promising targeted therapy for IgA nephropathy in mouse models.


Subject(s)
Glomerulonephritis, IGA , Animals , Disease Models, Animal , Female , Firmicutes , Glomerulonephritis, IGA/therapy , Humans , Immunoglobulin A/metabolism , Immunoglobulin G , Male , Mice , Receptors, Fc , Serine Endopeptidases
14.
Front Immunol ; 12: 676919, 2021.
Article in English | MEDLINE | ID: mdl-34594322

ABSTRACT

Introduction: Crescents, especially those found at a percentage greater than 50%, are often associated with rapid progression of kidney disease in IgA nephropathy (IgAN). The mechanism of crescents forming in IgAN is still unclear. In this study, we aimed to evaluate whether excess complement activation participates in the formation of crescents in IgAN. Methods: One hundred IgAN patients with various proportions of crescents-24 with 1%-24%, 27 with 25%-49%, 21 with 50%-74% 12 with more than 75%, and 16 without crescents-were included. Urinary concentrations of mannose-binding lectin (MBL), Bb, C4d, C3a, C5a, and soluble C5b-9 (sC5b-9) were measured at the time of biopsy. Receiver operating characteristic (ROC) curves were performed to evaluate predictive ability of renal survival for urine complement activation. In addition, historical C4d, C5b-9, and C3d were stained by immunohistochemistry. Results: IgAN patients with more than 50% crescent formation showed higher complement activation levels than the other patients (urinary C3a/creatinine (C3a/Cr): 6.7295 ng/mg, interquartile range (IQR) 1.4652-62.1086 ng/mg vs. 0.1055 ng/mg, IQR 0-1.4089 ng/mg; urinary C5a/Cr: 15.6202 ng/mg, 4.3127-66.7347 ng/mg vs. 0.3280 ng/mg, IQR 0.0859-2.4439 ng/mg; urinary sC5b-9/Cr: 98.6357 ng/mg, 8.8058-1,087.4578 ng/mg vs. 1.4262 ng/mg, 0.0916-11.0858 ng/mg, all p-values <0.001). The levels of urinary MBL and C4d representing lectin complement pathway showed a linear association with the proportion of crescents (r = 0.457 and 0.562, respectively, both p-values <0.001). Combined urine complement products could increase the predictive ability compared with crescents alone from 0.904 to 0.944 (p = 0.062) with borderline significance. Moreover, the glomerular C4d deposition rate elevated with the increase of proportions of crescents. Conclusion: Excess complement activation may be involved in the formation of crescents, especially diffuse crescent formation, in patients with IgAN. Urinary C4d correlated with the proportion of crescents and was a potential biomarker for disease monitoring in crescentic IgAN.


Subject(s)
Complement Activation , Complement System Proteins/urine , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/pathology , Severity of Illness Index , Adult , Biopsy , Female , Glomerulonephritis, IGA/blood , Glomerulonephritis, IGA/urine , Humans , Immunohistochemistry/methods , Kidney Glomerulus/pathology , Male , Mannose-Binding Lectin/urine , Middle Aged , Prognosis , Young Adult
15.
Clin J Am Soc Nephrol ; 16(11): 1652-1664, 2021 11.
Article in English | MEDLINE | ID: mdl-34607844

ABSTRACT

BACKGROUND AND OBJECTIVES: Poly-IgA immune complex formation and glomerular deposition play a key role in IgA nephropathy. Our study sought to develop a new methodology for one-step serologic detection of poly-IgA levels. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: A novel ELISA method using recombinant CD89 as a "capturing" probe was established for detecting poly-IgA immune complex in plasma. We applied semiquantitative measurements of these poly-IgA indices in patients recruited at Peking University First Hospital who had IgA nephropathy or other kidney disease types, as compared with healthy controls. The longitudinal trend of the poly-IgA index and the association with pathologic parameters and treatment responses were evaluated. Finally, we analyzed the molecular composition of poly-IgA complexes in patients by mass spectrometry. RESULTS: Recombinant CD89-mounted ELISA plates specifically captured plasma poly-IgA. The levels of poly-IgA immune complex (26.7 [interquartile range (IQR) 17.1-42.6] U/ml) in IgA nephropathy were significantly higher than those in healthy controls (15.5 [IQR 10.7-20.0] U/ml; P<0.001) or in controls with non-IgA nephropathy disease (14.8 [IQR 10.5-21.9] U/ml; P<0.001). Higher levels of poly-IgA immune complex were associated with lower eGFR and worse kidney outcome. Accuracy parameters and concordant statistics showed good discrimination between IgA nephropathy and healthy controls based on poly-IgA index levels (area under the curve [AUC], 0.78; 95% confidence interval [95% CI], 0.72 to 0.83; P<0.001), significantly outperforming galactose-deficient IgA1 levels (AUC, 0.70; P=0.05). Corticosteroid and immunosuppressant treatments lowered poly-IgA indices. After a recombinant CD89-directed workflow in conjunction with mass spectrometry, we also analyzed the molecular composition of IgA immune complex in patients with IgA nephropathy. CONCLUSIONS: Higher level of recombinant CD89-bound poly-IgA immune complex was associated with the severity of the disease and with treatment response to steroids and immunosuppressants.


Subject(s)
Glomerulonephritis, IGA/blood , Glomerulonephritis, IGA/drug therapy , Immunoglobulin A/metabolism , Adolescent , Adult , Antigens, CD , Area Under Curve , Case-Control Studies , Enzyme-Linked Immunosorbent Assay/methods , Female , Glomerular Filtration Rate , Glomerulonephritis, IGA/physiopathology , Glucocorticoids/therapeutic use , Humans , Immunosorbents , Immunosuppressive Agents/therapeutic use , Kidney Glomerulus , Male , Middle Aged , Receptors, Fc , Recombinant Proteins , Serologic Tests/methods , Severity of Illness Index , Young Adult
16.
Theranostics ; 11(18): 9118-9132, 2021.
Article in English | MEDLINE | ID: mdl-34522230

ABSTRACT

Rationale: A robust radiopharmaceutical has high uptake in the target and low retention in non-target tissues. However, traditional tracers for renal imaging that chemically chelate 99mTc are excreted through the renal route with transient resident time in the kidney. Following a rational design approach, we constructed a protein-based radiotracer, designated PBT-Fc, to sequentially bind tubular neonatal Fc-receptor and subsequently proximal tubular basement membrane for its targeted sequestration in kidney parenchyma. In this process, the tracer participates in physiologic glomerular filtration and tubular reabsorption while escaping lysosomal catabolism and urinary clearance. Methods: To specifically target renal receptors in navigating the urinary passage in the kidney, we produced a recombinant fusion protein with two separate functional parts: a polybasic PBT segment derived from human Vascular Endothelial Growth Factor and Fc segment of IgG1. The chimeric fusion of PBT-Fc was labeled with radionuclide 99mTc and tested in rodent models of kidney diseases. Planar scintigraphy and single-photon emission computerized tomography (SPECT) were performed to evaluate renal-specificity of the tracer. Results: When injected in mouse and rat, following a brief 10 - 15 min dynamic redistribution phase in circulation, ~ 95% of the [99mTc]-PBT-Fc signal was concentrated in the kidney and lasted for hours without urinary loss or surrounding tissue activities. Long-lasting tracer signals in the kidney cortex in conjunction with SPECT greatly augmented the image quality in detecting pathological lesions in a variety of disease models, including ischemic acute kidney injury, drug-induced renal toxicity, and chronic kidney disease from renin-angiotensin system (RAS) overactivation. Conclusion: Exclusive renal retention of the recombinant radiotracer greatly facilitated static-phase signal acquisition by SPECT and achieved submillimeter spatial resolution of kidney alternations in glomerular and tubular disease models.


Subject(s)
Kidney/diagnostic imaging , Single Photon Emission Computed Tomography Computed Tomography/methods , Animals , Histocompatibility Antigens Class I/metabolism , Kidney Function Tests/methods , Kidney Tubules/diagnostic imaging , Male , Mice , Mice, Inbred BALB C , Radioactive Tracers , Radioisotopes/pharmacokinetics , Radionuclide Imaging/methods , Radiopharmaceuticals/pharmacokinetics , Rats , Receptors, Fc/metabolism , Renal Reabsorption/physiology , Tomography, Emission-Computed, Single-Photon/methods
17.
JCI Insight ; 6(19)2021 10 08.
Article in English | MEDLINE | ID: mdl-34428184

ABSTRACT

IgA nephropathy is caused by deposition of circulatory IgA1 in the kidney. Hypogalactosylated IgA1 has the propensity to form poly-IgA aggregates that are prone to deposition. Herein, we purified poly-IgA from the plasma of patients with IgA nephropathy and showed that the complex is susceptible to reducing conditions, suggesting intermolecular disulfide connections between IgA units. We sought to find the cysteine residue(s) that form intermolecular disulfide. Naturally assembled dimeric IgA, also known as secretory IgA, involves a J chain subunit connected with 2 IgA1 molecules via their penultimate cysteine-471 residue on a "tailpiece" segment of IgA heavy chain. It is plausible that, with the absence of J chain, the cysteine residue of mono-IgA1 might aberrantly form a disulfide bond in poly-IgA formation. Mutagenesis confirmed that cysteine-471 is capable of promoting IgA aggregation. These discoveries prompted us to test thiol-based drugs for stabilizing cysteine. Specifically, the cystine-reducing drug cysteamine used for treatment of cystinosis showed a remarkable potency in preventing self-aggregation of IgA. When administrated to rat and mouse models of IgA nephropathy, cysteamine significantly reduced glomerular IgA deposition. Collectively, our results reveal a potentially novel molecular mechanism for aberrant formation of IgA aggregates, to which the repurposed cystinosis drug cysteamine was efficacious in preventing renal IgA deposition.


Subject(s)
Cysteine/metabolism , Glomerulonephritis, IGA/metabolism , Immunoglobulin A/metabolism , Kidney Glomerulus/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Cysteamine/pharmacology , Cysteine/drug effects , Cystine Depleting Agents/pharmacology , Disease Models, Animal , Humans , Immunoglobulin A/drug effects , Immunoglobulin J-Chains/metabolism , Kidney Glomerulus/drug effects , Mice , Rats
18.
Nephrol Dial Transplant ; 37(1): 33-41, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34152412

ABSTRACT

BACKGROUND: Immunoglobulin A1 (IgA1) O-glycosylation plays an important role in the pathogenesis of IgA nephropathy (IgAN). However, variations in IgA1 O-glycoforms have not been explored. We aimed to investigate the IgA1 O-glycoforms in the hinge region (HR) of polymeric IgA1 (pIgA1) and then evaluate the association between IgA1 O-glycoforms and crescent formation in IgAN. METHODS: The discovery cohort (Cohort 1) comprised 11 crescentic IgAN patients, 10 noncrescentic IgAN patients and 10 healthy controls and the validation cohort (Cohort 2) comprised 11 crescentic IgAN patients, 9 noncrescentic IgAN patients and 9 healthy controls. A total of 143 IgAN patients with different crescent percentages (Cohort 3) were also included. pIgA1 was purified from the plasma of the participants. The variation in O-glycoforms was evaluated by estimating the molecular weights of IgA1 hinge glycopeptides using reversed-phase liquid chromatography and tandem mass spectrometry under electron-transfer/higher-energy collision dissociation fragmentation mode. RESULTS: In the discovery cohort (Cohort 1), the number of N-acetylgalactosamine (GalNAc) bound to one HR was lower in IgAN patients. The proportions of GalNAc3 (defined as O-glycans bound to one HR at three sites) and GalNAc4 were highest in crescentic IgAN patients, followed by noncrescentic IgAN patients, and were lowest in healthy controls [GalNAc 3: 9.92 ± 3.37% versus 6.65 ± 1.53% versus 4.05 ± 1.24% (P < 0.001); GalNAc4: 45.91 ± 4.75% versus 41.13 ± 2.95% versus 40.98 ± 2.95% (P = 0.004), respectively]. The proportions of GalNAc5 and GalNAc6 were lowest in crescentic IgAN patients followed by noncrescentic IgAN patients and were highest in healthy controls [GalNAc5: 50.15 ± 4.27% versus 47.92 ± 4.09% versus 45.87 ± 3.79% (P = 0.028); GalNAc6: 6.58 ± 2.53% versus 6.04 ± 1.35% versus 4.65 ± 2.27% (P = 0.034), respectively]. These results were consistent in the validation cohort (Cohort 2). In another cohort with 143 patients with different crescent percentages (Cohort 3), the number of GalNAc in pIgA1 decreased with an increasing percentage of crescents. CONCLUSIONS: The number of GalNAc in IgA1 HRs was lower in IgAN patients, especially in crescentic IgAN patients, and may be associated with a severe IgAN phenotype.


Subject(s)
Glomerulonephritis, IGA , Glycosylation , Humans , Immunoglobulin A , Phenotype , Polysaccharides
20.
J Pathol ; 254(2): 159-172, 2021 06.
Article in English | MEDLINE | ID: mdl-33660264

ABSTRACT

IgA nephropathy (IgAN) is the most common type of glomerulonephritis worldwide, which follows a chronic but nonetheless highly variable course of progression. IgA immune complexes are the primary source of renal deposits in IgAN. Apart from the presence of granular IgA1 deposits in the glomerular mesangium and mesangial hypercellularity as common features, the detailed process of IgA1 deposition and clearance in the kidney remains unclear. We sought to examine the dynamics of IgA deposition and tissue plasticity in response to deposits including their intrarenal clearance. We followed a synthetic approach to produce a recombinant fusion between IgA Fc (rIgA) and a biotin tag, which was subsequently induced with streptavidin (SA) to form an oligomeric poly-IgA mimic. Both uninduced rIgA (mono-rIgA) and polymeric SA-rIgA (poly-rIgA) were injected intravenously into Wistar rats. Plasma IgA levels and renal and liver histology were examined in a time series. In contrast to mono-rIgA, this synthetic poly-rIgA analog formed renal deposits exclusively in the glomerulus and were mostly cleared in 3 h. However, repeated daily injections for 12 days caused long-lasting and stronger glomerular IgA deposition together with IgG and complement C3, in association with mesangial cell proliferation, matrix expansion, and variable degrees of albuminuria and hematuria that phenocopied IgAN. Ex vivo, poly-rIgA bound cultured mesangial cells and elicited cytokine production, in addition to activating plasma C3 that was consistent with the actions of IgA immune complexes in IgAN pathogenesis. Remarkably, the kidneys were able to reverse all pathologic manifestations and restore normal glomerular histology 2 weeks after injections were halted. The synthetic model showed the kinetics between the intricate balance of renal deposition and clearance, as well as glomerular plasticity towards healing. Together, the results revealed a priming effect of existing deposits in promoting stronger and longer-lasting IgA deposition to cause renal damage. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Complement C3/immunology , Glomerulonephritis, IGA/pathology , Glomerulonephritis/pathology , Immunoglobulin A/immunology , Albuminuria/immunology , Albuminuria/pathology , Animals , Disease Models, Animal , Glomerulonephritis/immunology , Glomerulonephritis, IGA/immunology , Hematuria/immunology , Hematuria/pathology , Humans , Immunoglobulin G/immunology , Kidney/immunology , Kidney/pathology , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Male , Mesangial Cells/immunology , Rats , Rats, Wistar , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...