Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Arthritis Res Ther ; 22(1): 95, 2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32345366

RESUMO

BACKGROUND: CD4+ T cells play a central role during the early stages of rheumatoid arthritis (RA), but to which extent they are required for the perpetuation of the disease is still not fully understood. The aim of the current study was to obtain conclusive evidence that T cells drive chronic relapsing arthritis. METHODS: We used the rat pristane-induced arthritis model, which accurately portrays the chronic relapsing-remitting disease course of RA, to examine the contribution of T cells to chronic arthritis. RESULTS: Rats subjected to whole-body irradiation and injected with CD4+ T cells from lymph nodes of pristane-injected donors developed chronic arthritis that lasted for more than 4 months, whereas T cells from the spleen only induced acute disease. Thymectomy in combination with irradiation enhanced the severity of arthritis, suggesting that sustained lymphopenia promotes T cell-driven chronic inflammation in this model. The ability of T cells to induce chronic arthritis correlated with their expression of Th17-associated transcripts, and while depletion of T cells in rats with chronic PIA led to transient, albeit significant, reduction in disease, neutralization of IL-17 resulted in almost complete and sustained remission. CONCLUSION: These findings show that, once activated, self-reactive T cells can sustain inflammatory responses for extended periods of time and suggest that such responses are promoted in the presence of IL-17.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Autoimunidade/imunologia , Linfócitos T CD4-Positivos/imunologia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/transplante , Doença Crônica , Feminino , Interleucina-17/imunologia , Masculino , Ratos
3.
Antioxid Redox Signal ; 30(4): 489-504, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29471681

RESUMO

AIMS: Human α1-microglobulin (A1M) is an endogenous reductase and radical- and heme-binding protein with physiological antioxidant protective functions. Recombinant human A1M (rA1M) has been shown to have therapeutic properties in animal models of preeclampsia, a pregnancy disease associated with oxidative stress. Recombinant A1M, however, lacks glycosylation, and shows lower solubility and stability than A1M purified from human plasma. The aims of this work were to (i) use site-directed mutagenesis to improve the physicochemical properties of rA1M, (ii) demonstrate that the physicochemically improved rA1M displays full in vitro cell protective effects as recombinant wild-type A1M (rA1M-wt), and (iii) show its therapeutic potential in vivo against acute kidney injury (AKI), another disease associated with oxidative stress. RESULTS: A novel recombinant A1M-variant (rA1M-035) with three amino acid substitutions was constructed, successfully expressed, and purified. rA1M-035 had improved solubility and stability compared with rA1M-wt, and showed intact in vitro heme-binding, reductase, antioxidation, and cell protective activities. Both rA1M-035 and rA1M-wt showed, for the first time, potential in vivo protective effects on kidneys using a mouse rhabdomyolysis glycerol injection model of AKI. INNOVATION: A novel recombinant A1M-variant, rA1M-035, was engineered. This protein showed improved solubility and stability compared with rA1M-wt, full in vitro functional activity, and potential protection against AKI in an in vivo rhabdomyolysis mouse model. CONCLUSION: The new rA1M-035 is a better drug candidate than rA1M-wt for treatment of AKI and preeclampsia in human patients.


Assuntos
Injúria Renal Aguda/complicações , Injúria Renal Aguda/terapia , alfa-Globulinas/metabolismo , Rabdomiólise/metabolismo , Injúria Renal Aguda/metabolismo , alfa-Globulinas/genética , Animais , Feminino , Humanos , Células K562 , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/metabolismo
4.
Biochim Biophys Acta ; 1864(1): 29-41, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26497278

RESUMO

BACKGROUND: α1-Microglobulin (A1M) is a reductase and radical scavenger involved in physiological protection against oxidative damage. These functions were previously shown to be dependent upon cysteinyl-, C34, and lysyl side-chains, K(92, 118,130). A1M binds heme and the crystal structure suggests that C34 and H123 participate in a heme binding site. We have investigated the involvement of these five residues in the interactions with heme. METHODS: Four A1M-variants were expressed: with cysteine to serine substitution in position 34, lysine to threonine substitutions in positions (92, 118, 130), histidine to serine substitution in position 123 and a wt without mutations. Heme binding was investigated by tryptophan fluorescence quenching, UV-Vis spectrophotometry, circular dichroism, SPR, electrophoretic migration shift, gel filtration, catalase-like activity and molecular simulation. RESULTS: All A1M-variants bound to heme. Mutations in C34, H123 or K(92, 118, 130) resulted in significant absorbance changes, CD spectral changes, and catalase-like activity, suggesting involvement of these side-groups in coordination of the heme-iron. Molecular simulation support a model with two heme-binding sites in A1M involving the mutated residues. Binding of the first heme induces allosteric stabilization of the structure predisposing for a better fit of the second heme. CONCLUSIONS: The results suggest that one heme-binding site is located in the lipocalin pocket and a second binding site between loops 1 and 4. Reactions with the hemes involve the side-groups of C34, K(92, 118, 130) and H123. GENERAL SIGNIFICANCE: The model provides a structural basis for the functional activities of A1M: heme binding activity of A1M.


Assuntos
alfa-Globulinas/química , Heme/química , Simulação de Dinâmica Molecular , Estrutura Terciária de Proteína , alfa-Globulinas/genética , alfa-Globulinas/metabolismo , Sítios de Ligação/genética , Western Blotting , Dicroísmo Circular , Heme/metabolismo , Humanos , Mutagênese Sítio-Dirigida/métodos , Mutação , Oxirredução , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Espectrometria de Fluorescência , Ressonância de Plasmônio de Superfície
5.
PLoS One ; 10(5): e0125499, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25955715

RESUMO

Preeclampsia is one of the most serious pregnancy-related diseases and clinically manifests as hypertension and proteinuria after 20 gestational weeks. The worldwide prevalence is 3-8% of pregnancies, making it the most common cause of maternal and fetal morbidity and mortality. Preeclampsia lacks an effective therapy, and the only "cure" is delivery. We have previously shown that increased synthesis and accumulation of cell-free fetal hemoglobin (HbF) in the placenta is important in the pathophysiology of preeclampsia. Extracellular hemoglobin (Hb) and its metabolites induce oxidative stress, which may lead to acute renal failure and vascular dysfunction seen in preeclampsia. The human endogenous protein, α1-microglobulin (A1M), removes cell-free heme-groups and induces natural tissue repair mechanisms. Exogenously administered A1M has been shown to alleviate the effects of Hb-induced oxidative stress in rat kidneys. Here we attempted to establish an animal model mimicking the human symptoms at stage two of preeclampsia by administering species-specific cell-free HbF starting mid-gestation until term, and evaluated the therapeutic effect of A1M on the induced symptoms. Female pregnant rabbits received HbF infusions i.v. with or without A1M every second day from gestational day 20. The HbF-infused animals developed proteinuria and a significantly increased glomerular sieving coefficient in kidney that was ameliorated by co-administration of A1M. Transmission electron microscopy analysis of kidney and placenta showed both intracellular and extracellular tissue damages after HbF-treatment, while A1M co-administration resulted in a significant reduction of the structural and cellular changes. Neither of the HbF-treated animals displayed any changes in blood pressure during pregnancy. In conclusion, infusion of cell-free HbF in the pregnant rabbits induced tissue damage and organ failure similar to those seen in preeclampsia, and was restored by co-administration of A1M. This study provides preclinical evidence supporting further examination of A1M as a potential new therapy for preeclampsia.


Assuntos
alfa-Globulinas/administração & dosagem , Hemoglobina Fetal/efeitos adversos , Glomérulos Renais/efeitos dos fármacos , Placenta/efeitos dos fármacos , Pré-Eclâmpsia/tratamento farmacológico , Proteinúria/tratamento farmacológico , alfa-Globulinas/metabolismo , Animais , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Hemoglobina Fetal/antagonistas & inibidores , Hemoglobina Fetal/metabolismo , Heme/antagonistas & inibidores , Heme/metabolismo , Humanos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Estresse Oxidativo/efeitos dos fármacos , Placenta/metabolismo , Placenta/patologia , Pré-Eclâmpsia/sangue , Pré-Eclâmpsia/induzido quimicamente , Pré-Eclâmpsia/patologia , Gravidez , Proteinúria/sangue , Proteinúria/induzido quimicamente , Proteinúria/patologia , Coelhos
6.
Front Physiol ; 5: 465, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25538624

RESUMO

BACKGROUND: Alpha-1-microglobulin (A1M), a small lipocalin protein found in plasma and tissues, has been identified as a heme and radical scavenger that may participate in the mitigation of toxicities caused by degradation of hemoglobin. The objective of this work was to investigate heme interactions with A1M in vitro using various analytical techniques and to optimize analytical methodology suitable for rapid evaluation of the ligand binding properties of recombinant A1M versions. METHODS: To examine heme binding properties of A1M we utilized UV/Vis absorption spectroscopy, visible circular dichroism (CD), catalase-like activity, migration shift electrophoresis, and surface plasmon resonance (SPR), which was specifically developed for the assessment of His-tagged A1M. RESULTS: The results of this study confirm that A1M is a heme binding protein that can accommodate heme at more than one binding site and/or in coordination with different amino acid residues depending upon heme concentration and ligand-to-protein molar ratio. UV/Vis titration of A1M with heme revealed an unusually large bathochromic shift, up to 38 nm, observed for heme binding to a primary binding site. UV/Vis spectroscopy, visible CD and catalase-like activity suggested that heme is accommodated inside His-tagged (tgA1M) and tagless A1M (ntA1M) in a rather similar fashion although the His-tag is very likely involved into coordination with iron of the heme molecule. SPR data indicated kinetic rate constants and equilibrium binding constants with KD values in a µM range. CONCLUSIONS: This study provided experimental evidence of the A1M heme binding properties by aid of different techniques and suggested an analytical methodology for a rapid evaluation of ligand-binding properties of recombinant A1M versions, also suitable for other His-tagged proteins.

7.
PLoS One ; 9(1): e86353, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24489717

RESUMO

Preeclampsia (PE) is a serious pregnancy complication that manifests as hypertension and proteinuria after the 20(th) gestation week. Previously, fetal hemoglobin (HbF) has been identified as a plausible causative factor. Cell-free Hb and its degradation products are known to cause oxidative stress and tissue damage, typical of the PE placenta. A1M (α1-microglobulin) is an endogenous scavenger of radicals and heme. Here, the usefulness of A1M as a treatment for PE is investigated in the pregnant ewe PE model, in which starvation induces PE symptoms via hemolysis. Eleven ewes, in late pregnancy, were starved for 36 hours and then treated with A1M (n = 5) or placebo (n = 6) injections. After injections, the ewes were re-fed and observed for additional 72 hours. They were monitored for blood pressure, proteinuria, blood cell distribution and clinical and inflammation markers in plasma. Before termination, the utero-placental circulation was analyzed with Doppler velocimetry and the kidney glomerular function was analyzed by Ficoll sieving. At termination, blood, kidney and placenta samples were collected and analyzed for changes in gene expression and tissue structure. The starvation resulted in increased amounts of the hemolysis marker bilirubin in the blood, structural damages to the placenta and kidneys and an increased glomerular sieving coefficient indicating a defect filtration barrier. Treatment with A1M ameliorated these changes without signs of side-effects. In conclusion, A1M displayed positive therapeutic effects in the ewe starvation PE model, and was well tolerated. Therefore, we suggest A1M as a plausible treatment for PE in humans.


Assuntos
alfa-Globulinas/metabolismo , Heme/toxicidade , Rim/efeitos dos fármacos , Rim/metabolismo , Placenta/efeitos dos fármacos , Placenta/metabolismo , Pré-Eclâmpsia/tratamento farmacológico , Pré-Eclâmpsia/metabolismo , alfa-Globulinas/genética , Animais , Feminino , Gravidez
8.
Antioxid Redox Signal ; 18(16): 2017-28, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23157686

RESUMO

AIMS: During cell death, energy-consuming cell degradation and recycling programs are performed. Maintenance of energy delivery during cell death is therefore crucial, but the mechanisms to keep the mitochondrial functions intact during these processes are poorly understood. We have investigated the hypothesis that the heme- and radical-binding ubiquitous protein α1-microglobulin (A1M) is involved in protection of the mitochondria against oxidative insult during cell death. RESULTS: Using blood cells, keratinocytes, and liver cells, we show that A1M binds with high affinity to apoptosis-induced cells and is localized to mitochondria. The mitochondrial Complex I subunit NDUFAB1 was identified as a major molecular target of the A1M binding. Furthermore, A1M was shown to inhibit the swelling of mitochondria, and to reverse the severely abrogated ATP-production of mitochondria when exposed to heme and reactive oxygen species (ROS). INNOVATION: Import of the radical- and heme-binding protein A1M from the extracellular compartment confers protection of the mitochondrial structure and function during cellular insult. CONCLUSION: A1M binds to a subunit of Complex I and has a role in assisting the mitochondria to maintain its energy delivery during cell death. A1M may also, at the same time, counteract and eliminate the ROS generated by the mitochondrial respiration to prevent oxidative damage to surrounding healthy tissue.


Assuntos
alfa-Globulinas/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Lipocalinas/metabolismo , Mitocôndrias/fisiologia , Animais , Células Cultivadas , Humanos , Camundongos , Mitocôndrias/metabolismo , Ligação Proteica
9.
Pregnancy Hypertens ; 3(2): 70-1, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26105866

RESUMO

INTRODUCTION: Previous gene expression analysis have identified fetal hemoglobin (HbF) as a plausible etiological factor in preeclampsia. Free hemoglobin and its degradation products, e.g. heme, are known to cause oxidative stress, tissue damage, and vaso-constriction, typical findings in preeclampsia. OBJECTIVE: To study alpha-1-microglobulin (A1M), an endogenous radical scavenger and heme-binder, as a potential treatment for preeclampsia using the pregnant ewe preeclampsia model. Free Hb and heme are known to take part in the pathology of this model and therefor well suited for evaluation of recombinant A1M as a therapy. METHODS: 11 pregnant ewes, at gestational age 125-131 days, were acclimatized for 36h and then starved for another 36h to induce preeclampsia symptoms. At the end of starvation period, they were treated either with placebo (n=6) or A1M injections (n=5). After injections, food was re-introduced and ewes further followed for 72h. The ewes were sacrificed the 6th day after beginning of acclimatization. Throughout the 6 days, the animals were monitored for blood pressure and different blood and urine parameters. Whole blood, kidney and placenta tissue samples were collected from the ewes. Gene expression analysis, blood analysis, histology and electron microscopy were used to evaluate the therapeutic effects of A1M. RESULTS: Starvation increased the amount of free heme in the blood. The ultrastructure of the placenta and kidney were damaged in a way similar to what previously have been described for PE. The glomeruli and the tubuli were damaged which was reflected by increased Ficol clearance and increased plasma creatinine levels. Treatment with A1M significantly normalized the kidney functions. The most profound changes on gene expression level were found in white blood cells in the starved animals. Starvation decreases mRNA expression for anti-oxidants such as CAT (P=0.04), SOD1 (P=0.008), SOD2 (1.8-fold) as well as angiogenetic factors such as VEGF (P=0.02) and HGF (1.6-fold). A1M treatment rescued the decreased expression of SOD2 (P=0.04) and HGF (2-fold). CONCLUSION: A1M is well tolerated and shows high potential as a treatment for PE-like symptoms in the pregnant ewe model for PE.

10.
Hum Mol Genet ; 18(24): 4689-98, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19759059

RESUMO

Mitochondria are organelles of all nucleated cells, and variations in mtDNA sequence affect a wide spectrum of human diseases. However, animal models for mtDNA-associated diseases are rare, making it challenging to explore mechanisms underlying the contribution of mitochondria. Here, we identify a polymorphism in the mitochondrial genome, G-to-T at position 7778, which results in an aspartic acid-to-tyrosine (D-Y) substitution in the fifth amino acid of the highly conserved N-terminus of ATP synthase 8 (ATP8). Using a series of conplastic strains we show that this polymorphism increases susceptibility to multiple autoimmune diseases, including collagen-induced arthritis, autoimmune diabetes, nephritis and autoimmune pancreatitis. In addition, it impairs reproductive performance in females, but only in the MRL/MpJ strain. We also demonstrate that the mtAtp8 polymorphism alters mitochondrial performance, increasing H(2)O(2) production and affecting mitochondrial structure. Functional analysis reveals that the polymorphism increase the CD4 T cell adaptive potential to an oxidative phosphorylation impaired condition. Our findings provide direct experimental evidence for the role of mitochondria in autoimmunity and reproduction.


Assuntos
Doenças Autoimunes/genética , DNA Mitocondrial/genética , Infertilidade Feminina/genética , Mitocôndrias/enzimologia , ATPases Mitocondriais Próton-Translocadoras/genética , Reprodução/genética , Sequência de Aminoácidos , Animais , Feminino , Genoma Mitocondrial , Peróxido de Hidrogênio/metabolismo , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Polimorfismo Genético
11.
Genome Res ; 19(1): 159-65, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19037013

RESUMO

Previous reports have demonstrated that the mtDNA of mouse common inbred strains (CIS) originated from a single female ancestor and that mtDNA mutations occurred during CIS establishment. This situation provides a unique opportunity to investigate the impact of individual mtDNA variations on complex traits in mammals. In this study, we compiled the complete mtDNA sequences of 52 mouse CIS. Phylogenetic analysis demonstrated that 50 of the 52 CIS descended from a single female Mus musculus domesticus mouse, and mtDNA mutations have accumulated in 26 of the CIS. We then generated conplastic strains on the C57BL/6J background for 12 mtDNA variants with one to three functional mtDNA mutations. We also generated conplastic strains for mtDNA variants of the four M. musculus subspecies, each of which contains hundreds of mtDNA variations. In total, a panel of conplastic strains was generated for 16 mtDNA variants. Phenotypic analysis of the conplastic strains demonstrated that mtDNA variations affect susceptibility to experimental autoimmune encephalomyelitis and anxiety-related behavior, which confirms that mtDNA variations affect complex traits. Thus, we have developed a unique genetic resource that will facilitate exploration of the biochemical and physiological roles of mitochondria in complex traits.


Assuntos
DNA Mitocondrial/genética , Variação Genética , Animais , Ansiedade/genética , Comportamento Animal , Encefalomielite Autoimune Experimental/genética , Feminino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Mutação , Fenótipo , Filogenia
12.
Curr Genomics ; 9(7): 494-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19506738

RESUMO

The identification of quantitative trait loci, QTL, in arthritis animal models is a straight forward process. However, to identify the underlying genes is a great challenge. One strategy frequently used, is to combine QTL analysis with genomic/proteomic screens. This has resulted in a number of publications where carefully performed genomic analyses present likely candidate genes for their respective QTL s. However, seldom the findings are reconnected to the QTL controlled phenotypes. In this review, we use our own data as an illustrative example that "very likely candidate genes" identified by genomic/proteomics is not necessarily the same as true QTL underlying genes.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...