Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Annu Rev Biomed Eng ; 25: 1-21, 2023 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-37289555

RESUMO

Hemolysis (i.e., red blood cell lysis) can increase circulatory levels of cell-free hemoglobin (Hb) and its degradation by-products, namely heme (h) and iron (Fe). Under homeostasis, minor increases in these three hemolytic by-products (Hb/h/Fe) are rapidly scavenged and cleared by natural plasma proteins. Under certain pathophysiological conditions, scavenging systems become overwhelmed, leading to the accumulation of Hb/h/Fe in the circulation. Unfortunately, these species cause various side effects such as vasoconstriction, hypertension, and oxidative organ damage. Therefore, various therapeutics strategies are in development, ranging from supplementation with depleted plasma scavenger proteins to engineered biomimetic protein constructs capable of scavenging multiple hemolytic species. In this review, we briefly describe hemolysis and the characteristics of the major plasma-derived protein scavengers of Hb/h/Fe. Finally, we present novel engineering approaches designed to address the toxicity of these hemolytic by-products.


Assuntos
Heme , Hemólise , Humanos , Heme/metabolismo , Hemólise/fisiologia , Ferro , Haptoglobinas/metabolismo , Haptoglobinas/uso terapêutico , Hemoglobinas/metabolismo
2.
Blood ; 140(17): 1837-1844, 2022 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-35660854

RESUMO

During hemolysis, erythrophagocytes dispose damaged red blood cells. This prevents the extracellular release of hemoglobin, detoxifies heme, and recycles iron in a linked metabolic pathway. Complementary to this process, haptoglobin and hemopexin scavenge and shuttle the red blood cell toxins hemoglobin and heme to cellular clearance. Pathological hemolysis outpaces macrophage capacity and scavenger synthesis across a diversity of diseases. This imbalance leads to hemoglobin-driven disease progression. To meet a void in treatment options, scavenger protein-based therapeutics are in clinical development.


Assuntos
Hemólise , Hemopexina , Humanos , Hemoglobinas/metabolismo , Haptoglobinas/metabolismo , Haptoglobinas/uso terapêutico , Heme/metabolismo
3.
Rheumatol Int ; 44(3): 483-495, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37847388

RESUMO

According to the Assessment of SpondyloArthritis International Society-European Alliance of Associations for Rheumatology (ASAS-EULAR) recommendations for the management of axial spondyloarthritis (axSpA), patients should undergo at least two courses of non-steroidal anti-inflammatory drugs (NSAIDs) therapy. In our study, we enrolled axSpA patients both at onset and in a flare who had already been treated with NSAIDs ineffectively. Subsequently, according to the recommendations, they received modified NSAID treatment as another attempt to the first-line drug therapy and were monitored from there. We aimed to identify risk factors for treatment failure after 4 weeks (Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) score ≥ 4) especially amongst zonulin and haptoglobin concentrations, and haptoglobin polymorphism. Treatment failure was observed in 71% of patients, and the following variables were contributed for occurrence of this state: higher zonulin levels, ankylosing spondylitis, X-ray sacroiliitis, magnetic resonance imaging sacroiliitis, long duration of symptoms, high BASDAI, and high value of spinal pain intensity on visual analogue scale. In addition, the following positive correlations were found: haptoglobin concentration with C-reactive protein (r = 0.56; p = 0.0004), and erythrocyte sedimentation rate (r = 0.62; p < 0.0001), as well as between zonulin levels and white blood count (r = 0.5; p = 0.0003). The results of the study presented the identified factors related to the standard treatment failure in axSpA, amongst them zonulin levels. They might be applied to point out the patients for whom the search for a more appropriate method of treatment should be considered.


Assuntos
Precursores de Proteínas , Sacroileíte , Espondilartrite , Espondilite Anquilosante , Humanos , Espondilite Anquilosante/tratamento farmacológico , Espondilite Anquilosante/genética , Espondilite Anquilosante/diagnóstico , Haptoglobinas/genética , Haptoglobinas/uso terapêutico , Sacroileíte/diagnóstico , Anti-Inflamatórios não Esteroides/uso terapêutico , Espondilartrite/diagnóstico , Falha de Tratamento
4.
Stroke ; 54(7): 1930-1942, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37232189

RESUMO

Aneurysmal subarachnoid hemorrhage (aSAH) is a devastating form of stroke frequently affecting young to middle-aged adults, with an unmet need to improve outcome. This special report focusses on the development of intrathecal haptoglobin supplementation as a treatment by reviewing current knowledge and progress, arriving at a Delphi-based global consensus regarding the pathophysiological role of extracellular hemoglobin and research priorities for clinical translation of hemoglobin-scavenging therapeutics. After aneurysmal subarachnoid hemorrhage, erythrocyte lysis generates cell-free hemoglobin in the cerebrospinal fluid, which is a strong determinant of secondary brain injury and long-term clinical outcome. Haptoglobin is the body's first-line defense against cell-free hemoglobin by binding it irreversibly, preventing translocation of hemoglobin into the brain parenchyma and nitric oxide-sensitive functional compartments of cerebral arteries. In mouse and sheep models, intraventricular administration of haptoglobin reversed hemoglobin-induced clinical, histological, and biochemical features of human aneurysmal subarachnoid hemorrhage. Clinical translation of this strategy imposes unique challenges set by the novel mode of action and the anticipated need for intrathecal drug administration, necessitating early input from stakeholders. Practising clinicians (n=72) and scientific experts (n=28) from 5 continents participated in the Delphi study. Inflammation, microvascular spasm, initial intracranial pressure increase, and disruption of nitric oxide signaling were deemed the most important pathophysiological pathways determining outcome. Cell-free hemoglobin was thought to play an important role mostly in pathways related to iron toxicity, oxidative stress, nitric oxide, and inflammation. While useful, there was consensus that further preclinical work was not a priority, with most believing the field was ready for an early phase trial. The highest research priorities were related to confirming haptoglobin's anticipated safety, individualized versus standard dosing, timing of treatment, pharmacokinetics, pharmacodynamics, and outcome measure selection. These results highlight the need for early phase trials of intracranial haptoglobin for aneurysmal subarachnoid hemorrhage, and the value of early input from clinical disciplines on a global scale during the early stages of clinical translation.


Assuntos
Hemorragia Subaracnóidea , Adulto , Pessoa de Meia-Idade , Humanos , Animais , Camundongos , Ovinos , Hemorragia Subaracnóidea/tratamento farmacológico , Hemorragia Subaracnóidea/complicações , Haptoglobinas/uso terapêutico , Consenso , Óxido Nítrico , Inflamação/complicações , Hemoglobinas
5.
J Neurochem ; 167(1): 90-103, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37702203

RESUMO

During subarachnoid haemorrhage, a blood clot forms in the subarachnoid space releasing extracellular haemoglobin (Hb), which causes oxidative damage and cell death in surrounding tissues. High rates of disability and cognitive decline in SAH survivors are attributed to loss of neurons and functional connections during secondary brain injury. Haptoglobin sequesters Hb for clearance, but this scavenging system is overwhelmed after a haemorrhage. Whilst exogenous haptoglobin application can attenuate cytotoxicity of Hb in vitro and in vivo, the functional effects of sub-lethal Hb concentrations on surviving neurons and whether cellular function can be protected with haptoglobin treatment remain unclear. Here we use cultured neurons to investigate neuronal health and function across a range of Hb concentrations to establish the thresholds for cellular damage and investigate synaptic function. Hb impairs ATP concentrations and cytoskeletal structure. At clinically relevant but sub-lethal Hb concentrations, we find that synaptic AMPAR-driven currents are reduced, accompanied by a reduction in GluA1 subunit expression. Haptoglobin co-application can prevent these deficits by scavenging free Hb to reduce it to sub-threshold concentrations and does not need to be present at stoichiometric amounts to achieve efficacy. Haptoglobin itself does not impair measures of neuronal health and function at any concentration tested. Our data highlight a role for Hb in modifying synaptic function in surviving neurons, which may link to impaired cognition or plasticity after SAH and support the development of haptoglobin as a therapy for subarachnoid haemorrhage.


Assuntos
Lesões Encefálicas , Hemorragia Subaracnóidea , Humanos , Haptoglobinas/farmacologia , Haptoglobinas/uso terapêutico , Hemorragia Subaracnóidea/metabolismo , Hemoglobinas/farmacologia , Hemoglobinas/uso terapêutico , Neurônios/metabolismo , Lesões Encefálicas/metabolismo
6.
Medicina (Kaunas) ; 59(7)2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37476546

RESUMO

Colonic inflammatory bowel disease (IBD) encompasses ulcerative colitis (UC) and Crohn's colitis (CC). Patients with IBD are at increased risk for colitis-associated colorectal cancer (CACRC) compared to the general population. CACRC is preceded by IBD, characterized by highly heterogenous, pharmacologically incurable, pertinacious, worsening, and immune-mediated inflammatory pathologies of the colon and rectum. The molecular and immunological basis of CACRC is highly correlated with the duration and severity of inflammation, which is influenced by the exogenous free hemoglobin alpha chain (HbαC), a byproduct of infiltrating immune cells; extravasated erythrocytes; and macrophage erythrophagocytosis. The exogenous free HbαC prompts oxygen free radical-arbitrated DNA damage (DNAD) through increased cellular reactive oxygen species (ROS), which is exacerbated by decreased tissue antioxidant defenses. Mitigation of the Fenton Reaction via pharmaceutical therapy would attenuate ROS, promote apoptosis and DNAD repair, and subsequently prevent the incidence of CACRC. Three pharmaceutical options that attenuate hemoglobin toxicity include haptoglobin, deferoxamine, and flavonoids (vitamins C/E). Haptoglobin's clearance rate from plasma is inversely correlated with its size; the smaller the size, the faster the clearance. Thus, the administration of Hp1-1 may prove to be beneficial. Further, deferoxamine's hydrophilic structure limits its ability to cross cell membranes. Finally, the effectiveness of flavonoids, natural herb antioxidants, is associated with the high reactivity of hydroxyl substituents. Multiple analyses are currently underway to assess the clinical context of CACRC and outline the molecular basis of HbαC-induced ROS pathogenesis by exposing colonocytes and/or colonoids to HbαC. The molecular immunopathogenesis pathways of CACRC herein reviewed are broadly still not well understood. Therefore, this timely review outlines the molecular and immunological basis of disease pathogenesis and pharmaceutical intervention as a protective measure for CACRC.


Assuntos
Neoplasias Colorretais , Doenças Inflamatórias Intestinais , Linfo-Histiocitose Hemofagocítica , Humanos , Antioxidantes , Desferroxamina/uso terapêutico , Eritrócitos/metabolismo , Eritrócitos/patologia , Haptoglobinas/uso terapêutico , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/uso terapêutico
7.
Neurosurg Rev ; 43(5): 1273-1288, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31493061

RESUMO

Delayed cerebral ischaemia (DCI) after aneurysmal subarachnoid haemorrhage (aSAH) is a major cause of mortality and morbidity. The pathophysiology of DCI after aSAH is thought to involve toxic mediators released from lysis of red blood cells within the subarachnoid space, including free haemoglobin and haem. Haptoglobin and hemopexin are endogenously produced acute phase proteins that are involved in the clearance of these toxic mediators. The aim of this review is to investigate the pathophysiological mechanisms involved in DCI and the role of both endogenous as well as exogenously administered haptoglobin and hemopexin in the prevention of DCI.


Assuntos
Isquemia Encefálica/etiologia , Isquemia Encefálica/prevenção & controle , Haptoglobinas/uso terapêutico , Hemopexina/uso terapêutico , Hemorragia Subaracnóidea/complicações , Humanos
8.
Transfusion ; 59(5): 1648-1650, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30702749

RESUMO

BACKGROUND: Drug-induced hemolytic anemia is a rare and potentially fatal complication of drug treatment. Specific laboratory tests are crucial to confirm the diagnosis. CASE REPORT: A 38-year-old woman on treatment with dimethyl fumarate for multiple sclerosis presented with a 7-day history of weakness and fatigue. Laboratory tests revealed profound hemolytic anemia with hemoglobin levels of 4.7 g/dL (reference, 12.5-16.0), decreased haptoglobin, increased reticulocyte count, and increased indirect bilirubin. A first direct antiglobulin test was negative. The patient was started on prednisone 1 mg/kg/day, and dimethyl fumarate was withdrawn. A blood sample was drawn on Day 7 and sent to a reference laboratory. A direct antiglobulin test performed 7 days later was positive. Furthermore, an indirect antiglobulin test was positive only in the presence of the drug. RESULTS: The patient did not receive a blood transfusion, recovered clinically during the following days, and was discharged on Day 7. On Day 36, the patient's RBCs had normalized. She was changed to another disease-modifying treatment for her multiple sclerosis, and at 10-month follow-up she remained stable without any notable adverse effects. CONCLUSION: This case describes the first report of a dimethyl fumarate-induced hemolytic anemia. Laboratory results should always be interpreted within the clinical context. Specific laboratory expertise is often needed, given the complexity of the field.


Assuntos
Anemia Hemolítica/induzido quimicamente , Fumarato de Dimetilo/toxicidade , Adulto , Transfusão de Sangue , Teste de Coombs , Feminino , Haptoglobinas/uso terapêutico , Humanos , Esclerose Múltipla/metabolismo , Prednisona/metabolismo
9.
Transfusion ; 59(12): 3628-3638, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31639229

RESUMO

BACKGROUND: During sepsis, higher plasma cell-free hemoglobin (CFH) levels portend worse outcomes. In sepsis models, plasma proteins that bind CFH improve survival. In our canine antibiotic-treated Staphylococcus aureus pneumonia model, with and without red blood cell (RBC) exchange transfusion, commercial human haptoglobin (Hp) concentrates bound and compartmentalized CFH intravascularly, increased CFH clearance, and lowered iron levels, improving shock, lung injury, and survival. We now investigate in our model how very high CFH levels and treatment time affect Hp's beneficial effects. MATERIALS AND METHODS: Two separate canine pneumonia sepsis Hp studies were undertaken: one with exchange transfusion of RBCs after prolonged storage to raise CFH to very high levels and another with rapidly lethal sepsis alone to shorten time to treat. All animals received continuous standard intensive care unit supportive care for 96 hours. RESULTS: Older RBCs markedly elevated plasma CFH levels and, when combined with Hp therapy, created supraphysiologic CFH-Hp complexes that did not increase CFH or iron clearance or improve lung injury and survival. In a rapidly lethal bacterial challenge model without RBC transfusion, Hp binding did not increase clearance of complexes or iron or show benefits seen previously in the less lethal model. DISCUSSION: High-level CFH-Hp complexes may impair clearance mechanisms and eliminate Hp's beneficial effect during sepsis. Rapidly lethal sepsis narrows the therapeutic window for CFH and iron clearance, also decreasing Hp's beneficial effects. In designing clinical trials, dosing and kinetics may be critical factors if Hp infusion is used to treat sepsis.


Assuntos
Haptoglobinas/uso terapêutico , Hemoglobinas/metabolismo , Pneumonia Estafilocócica/tratamento farmacológico , Pneumonia Estafilocócica/metabolismo , Choque Séptico/tratamento farmacológico , Choque Séptico/metabolismo , Animais , Modelos Animais de Doenças , Cães , Transfusão de Eritrócitos , Pneumonia Estafilocócica/terapia , Sepse/tratamento farmacológico , Sepse/metabolismo , Sepse/terapia , Choque Séptico/terapia
10.
J Intern Med ; 282(1): 76-93, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28464519

RESUMO

BACKGROUND: Extra-corpuscular haemoglobin is an endogenous factor enhancing inflammatory tissue damage, a process counteracted by the haemoglobin-binding plasma protein haptoglobin composed of alpha and beta subunits connected by disulfide bridges. Recent studies established that haptoglobin also binds and sequesters another pro-inflammatory mediator, HMGB1, via triggering CD163 receptor-mediated anti-inflammatory responses involving heme oxygenase-1 expression and IL-10 release. The molecular mechanism underlying haptoglobin-HMGB1 interaction remains poorly elucidated. METHODS: Haptoglobin ß subunits were tested for HMGB1-binding properties, as well as efficacy in animal models of sterile liver injury (induced by intraperitoneal acetaminophen administration) or infectious peritonitis (induced by cecal ligation and puncture, CLP, surgery) using wild-type (C57BL/6) or haptoglobin gene-deficient mice. RESULTS: Structural-functional analysis demonstrated that the haptoglobin ß subunit recapitulates the HMGB1-binding properties of full-length haptoglobin. Similar to HMGB1-haptoglobin complexes, the HMGB1-haptoglobin ß complexes also elicited anti-inflammatory effects via CD163-mediated IL-10 release and heme oxygenase-1 expression. Treatment with haptoglobin ß protein conferred significant protection in mouse models of polymicrobial sepsis as well as acetaminophen-induced liver injury, two HMGB1-dependent inflammatory conditions. CONCLUSIONS: Haptoglobin ß protein offers a novel therapeutic approach to fight against various inflammatory diseases caused by excessive HMGB1 release.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Proteína HMGB1/metabolismo , Haptoglobinas/metabolismo , Sepse/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/terapia , Citocinas/metabolismo , Modelos Animais de Doenças , Proteína HMGB1/antagonistas & inibidores , Haptoglobinas/uso terapêutico , Heme Oxigenase-1/biossíntese , Heme Oxigenase-1/metabolismo , Humanos , Interleucina-10/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Sepse/terapia
11.
Crit Care ; 21(1): 245, 2017 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-28946897

RESUMO

BACKGROUND: Intravascular haemolysis has been associated with acute kidney injury (AKI) in different clinical settings (cardiac surgery, sickle cell disease). Haemolysis occurs frequently in critically ill burn patients. The aim of this study was to assess the predictive value of haptoglobin at admission to predict major adverse kidney events (MAKE) and AKI in critically ill burn patients. METHODS: We conducted a retrospective, single-centre cohort study in a burn critical care unit in a tertiary centre, including all consecutive severely burned patients (total burned body surface > 20% and/or shock and/or mechanical ventilation at admission) from January 2012 to April 2017 with a plasmatic haptoglobin dosage at admission. RESULTS: A total of 130 patients were included in the analysis. Their mean age was 49 (34-62) years, their median total body surface area burned was 29% (15-51%) and the intensive care unit (ICU) mortality was 25%. Early haemolysis was defined as an undetectable plasmatic haptoglobin at admission. We used logistic regression to identify MAKE and AKI risk factors. In multivariate analysis, undetectable haptoglobin was associated with MAKE and AKI (respectively, OR 6.33, 95% CI 2.34-16.45, p < 0.001; OR 8.32, 95% CI 2.86-26.40, p < 0.001). CONCLUSIONS: Undetectable plasmatic haptoglobin at ICU admission is an independent risk factor for MAKE and AKI in critically ill burn patients. This study provides a rationale for biomarker-guided therapy using haptoglobin in critically ill burn patients.


Assuntos
Injúria Renal Aguda/etiologia , Queimaduras/complicações , Haptoglobinas/análise , Haptoglobinas/farmacologia , Injúria Renal Aguda/mortalidade , Adulto , Queimaduras/metabolismo , Queimaduras/mortalidade , Estudos de Coortes , Creatinina/análise , Creatinina/sangue , Feminino , Haptoglobinas/uso terapêutico , Humanos , Unidades de Terapia Intensiva/organização & administração , Masculino , Pessoa de Meia-Idade , Paris , Respiração Artificial/efeitos adversos , Estudos Retrospectivos , Fatores de Risco , Estatísticas não Paramétricas
12.
Blood ; 121(8): 1276-84, 2013 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-23264591

RESUMO

Hemolysis occurs in many hematologic and nonhematologic diseases. Extracellular hemoglobin (Hb) has been found to trigger specific pathophysiologies that are associated with adverse clinical outcomes in patients with hemolysis, such as acute and chronic vascular disease, inflammation, thrombosis, and renal impairment. Among the molecular characteristics of extracellular Hb, translocation of the molecule into the extravascular space, oxidative and nitric oxide reactions, hemin release, and molecular signaling effects of hemin appear to be the most critical. Limited clinical experience with a plasma-derived haptoglobin (Hp) product in Japan and more recent preclinical animal studies suggest that the natural Hb and the hemin-scavenger proteins Hp and hemopexin have a strong potential to neutralize the adverse physiologic effects of Hb and hemin. This includes conditions that are as diverse as RBC transfusion, sickle cell disease, sepsis, and extracorporeal circulation. This perspective reviews the principal mechanisms of Hb and hemin toxicity in different disease states, updates how the natural scavengers efficiently control these toxic moieties, and explores critical issues in the development of human plasma-derived Hp and hemopexin as therapeutics for patients with excessive intravascular hemolysis.


Assuntos
Haptoglobinas/uso terapêutico , Doenças Hematológicas/sangue , Doenças Hematológicas/tratamento farmacológico , Hemólise/efeitos dos fármacos , Hemólise/fisiologia , Hemopexina/uso terapêutico , Animais , Haptoglobinas/metabolismo , Hemina/metabolismo , Hemoglobinas/metabolismo , Hemopexina/metabolismo , Humanos
13.
Cells ; 11(19)2022 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-36230933

RESUMO

Intracerebral hemorrhage (ICH) is recognized as a severe clinical problem lacking effective treatment. High mobility group box-1 (HMGB1) exhibits inflammatory cytokine-like activity once released into the extracellular space from the nuclei. We previously demonstrated that intravenous injection of rat anti-HMGB1 monoclonal antibody (mAb) remarkably ameliorated brain injury in a rat ICH model. Therefore, we developed a humanized anti-HMGB1 mAb (OKY001) for clinical use. The present study examined whether and how the humanized anti-HMGB1 mAb ameliorates ICH injury in common marmosets. The results show that administration of humanized anti-HMGB1 mAb inhibited HMGB1 release from the brain into plasma, in association with a decrease of 4-hydroxynonenal (4-HNE) accumulation and a decrease in cerebral iron deposition. In addition, humanized anti-HMGB1 mAb treatment resulted in a reduction in brain injury volume at 12 d after ICH induction. Our in vitro experiment showed that recombinant HMGB1 inhibited hemoglobin uptake by macrophages through CD163 in the presence of haptoglobin, suggesting that the release of excess HMGB1 from the brain may induce a delay in hemoglobin scavenging, thereby allowing the toxic effects of hemoglobin, heme, and Fe2+ to persist. Finally, humanized anti-HMGB1 mAb reduced body weight loss and improved behavioral performance after ICH. Taken together, these results suggest that intravenous injection of humanized anti-HMGB1 mAb has potential as a novel therapeutic strategy for ICH.


Assuntos
Lesões Encefálicas , Callithrix , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Lesões Encefálicas/tratamento farmacológico , Hemorragia Cerebral/tratamento farmacológico , Citocinas , Proteína HMGB1/imunologia , Haptoglobinas/uso terapêutico , Heme , Ferro , Ratos , Ratos Wistar
14.
PLoS One ; 17(12): e0279416, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36542633

RESUMO

BACKGROUND: Patients with advanced fibrosis are at risk for developing hepatocellular carcinoma (HCC) even after hepatitis C virus (HCV) elimination. We previously reported that serum fucosylated haptoglobin (Fuc-Hp) levels increase as the disease progresses from chronic hepatitis to cirrhosis and then HCC. However, it remains unclear whether serum Fuc-Hp levels can stratify the risk of HCC occurrence after a sustained virological response (SVR) is achieved with direct-acting antivirals (DAAs) in patients with advanced liver fibrosis. METHODS: Among 3,550 patients with chronic hepatitis C treated with DAAs at Osaka University Hospital and related hospitals, the stored sera of 140 patients who were diagnosed with F3 or F4 by liver biopsy before DAA treatment, achieved SVR, and had no history of HCC were available at both baseline and the end of treatment (EOT). We measured the Fuc-Hp levels in these samples. RESULTS: The median serum levels of Fuc-Hp at EOT were significantly lower than those at baseline. During the 54.4-month follow-up period, 16 of 140 patients developed HCC. Multivariate Cox proportional hazards analysis revealed that high Fuc-Hp at EOT, high body mass index (BMI), and low albumin at EOT were independent risk factors for HCC occurrence. Patients with all three factors-high Fuc-Hp, high BMI, and low albumin-had a higher incidence of HCC than patients without these factors. CONCLUSIONS: High serum Fuc-Hp levels at EOT were an independent risk factor for HCC occurrence after SVR. Combined with BMI and albumin, Fuc-Hp can stratify the risk of HCC occurrence among those with advanced fibrosis.


Assuntos
Carcinoma Hepatocelular , Hepatite C Crônica , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patologia , Antivirais/uso terapêutico , Neoplasias Hepáticas/patologia , Hepacivirus , Haptoglobinas/uso terapêutico , Hepatite C Crônica/complicações , Hepatite C Crônica/tratamento farmacológico , Cirrose Hepática/diagnóstico , Resposta Viral Sustentada
15.
Int J Hematol ; 116(5): 812-818, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35790695

RESUMO

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


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Síndrome Hemolítico-Urêmica , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Púrpura Trombocitopênica Trombótica , Masculino , Humanos , Criança , Fator H do Complemento/genética , Fator H do Complemento/uso terapêutico , Hemólise , Haptoglobinas/uso terapêutico , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/patologia , Síndrome Hemolítico-Urêmica/terapia , Púrpura Trombocitopênica Trombótica/terapia , Proteínas do Sistema Complemento , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Síndrome Hemolítico-Urêmica Atípica/genética
16.
Dtsch Med Wochenschr ; 147(19): 1243-1250, 2022 09.
Artigo em Alemão | MEDLINE | ID: mdl-36126922

RESUMO

Autoimmune haemolytic anemia (AIHA) is defined as the immune-mediated destruction of red blood cells. In most cases, antibodies that target surface antigens on erythrocytes lead to their premature degradation in the spleen or, less commonly, in the liver. The term includes a heterogenous group of diseases, which differ largely in pathophysiology and treatment. The two most common entities are warm AIHA and cold AIHA. Diagnostic testing involves the analysis of haemolytic markers like lactate dehydrogenase, haptoglobin and unconjugated bilirubin as well as a hemoglobin and reticulocytes. In case of a haemolytic anemia, further testing like a blood smear and a direct antiglobulin test should follow. As diagnostic testing and treatment of AIHA are complex, affected patients should always be referred to a hematologist.In warm AIHA, mainly IgG autoantibodies bind to their antigen on the erythrocyte surface at body temperature, leading to their premature destruction in the spleen. First line treatment options include the administration of steroids which mitigate the destruction of red blood cells by macrophages in the spleen. In contrast, IgM autoantibodies in cold AIHA lead to intravasal agglutination of erythrocytes and complement activation. The IgM antibodies have their highest affinity below body temperature which is why patients experience symptoms mainly in cold-exposed body areas. Although the IgM antibodies dissolve at body temperature, the complement-loaded erythrocytes are destroyed in the liver. Therapeutic options include protection from cold and immunosuppressive agents or complement inhibition.


Assuntos
Anemia Hemolítica Autoimune , Haptoglobinas , Anemia Hemolítica Autoimune/tratamento farmacológico , Anemia Hemolítica Autoimune/terapia , Antígenos de Superfície/uso terapêutico , Autoanticorpos , Bilirrubina/uso terapêutico , Haptoglobinas/uso terapêutico , Humanos , Imunoglobulina G/uso terapêutico , Imunoglobulina M/uso terapêutico , Imunossupressores/uso terapêutico , Lactato Desidrogenases
17.
Acta Neurochir Suppl ; 111: 107-12, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21725739

RESUMO

After intracerebral hemorrhage (ICH), hemoglobin (Hb) that is released from erythrocytes within the brain hematoma is highly cytotoxic and leads to severe brain edema and direct neuronal damage. Therefore, neutralization of Hb could represent an important target for reducing the secondary injury after ICH. Haptoglobin (Hp), an endogenous Hb-binding protein in blood plasma, is found in this study to be upregulated in the hematoma-affected brain after ICH. Both in vivo and in vitro studies indicate that Hp upregulation is primarily mediated by oligodendrocytes. Hp acts as a secretory protein capable of neutralizing the cell-free Hb. We also found in an "ICH-like" injury that Hp-KO mice show the most severe brain injury and neurological deficits, whereas Hp-Tg mice are the most resistant to ICH injury, suggesting that a higher Hp level is associated with the increased resistance of animals to hemolytic product-mediated brain injury after ICH. We conclude that brain-derived Hp plays a cytoprotective role after ICH, and Hp may represent a new potential therapeutic target for management of ICH.


Assuntos
Lesões Encefálicas/etiologia , Lesões Encefálicas/prevenção & controle , Hemorragia Cerebral/complicações , Citoproteção , Haptoglobinas/uso terapêutico , Animais , Lesões Encefálicas/patologia , Células Cultivadas , Hemorragia Cerebral/patologia , Modelos Animais de Doenças , Embrião de Mamíferos , Regulação da Expressão Gênica/fisiologia , Haptoglobinas/deficiência , Haptoglobinas/metabolismo , L-Lactato Desidrogenase/metabolismo , Camundongos , Camundongos Knockout , Proteína Básica da Mielina/metabolismo , Neuroglia/metabolismo , Exame Neurológico , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley
18.
Trends Mol Med ; 26(7): 683-697, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32589936

RESUMO

Hemolysis and accumulation of cell-free hemoglobin (Hb) in the circulation or in confined tissue compartments such as the subarachnoid space is an important driver of disease. Haptoglobin is the Hb binding and clearance protein in human plasma and an efficient antagonist of Hb toxicity resulting from physiological red blood cell turnover. However, endogenous concentrations of haptoglobin are insufficient to provide protection against Hb-driven disease processes in conditions such as sickle cell anemia, sepsis, transfusion reactions, medical-device associated hemolysis, or after a subarachnoid hemorrhage. As a result, there is increasing interest in developing haptoglobin therapeutics to target 'toxic' cell-free Hb exposures. Here, we discuss key concepts of Hb toxicity and provide a perspective on the use of haptoglobin as a therapeutic protein.


Assuntos
Haptoglobinas/farmacologia , Haptoglobinas/uso terapêutico , Hemoglobinas/toxicidade , Anemia Falciforme/tratamento farmacológico , Animais , Eritrócitos/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Humanos , Sepse/tratamento farmacológico , Reação Transfusional/tratamento farmacológico
19.
Semin Nephrol ; 40(2): 148-159, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32303278

RESUMO

Sepsis is a heterogeneous clinical syndrome that is complicated commonly by acute kidney injury (sepsis-AKI). Currently, no approved pharmacologic therapies exist to either prevent sepsis-AKI or to treat sepsis-AKI once it occurs. A growing body of evidence supports a connection between red blood cell biology and sepsis-AKI. Increased levels of circulating cell-free hemoglobin (CFH) released from red blood cells during hemolysis are common during sepsis and can contribute to sepsis-AKI through several mechanisms including tubular obstruction, nitric oxide depletion, oxidative injury, and proinflammatory signaling. A number of potential pharmacologic therapies targeting CFH in sepsis have been identified including haptoglobin, hemopexin, and acetaminophen, and early phase clinical trials have suggested that acetaminophen may have beneficial effects on lipid peroxidation and kidney function in patients with sepsis. Bedside measurement of CFH levels may facilitate predictive enrichment for future clinical trials of CFH-targeted therapeutics. However, rapid and reliable bedside tests for plasma CFH will be required for such trials to move forward.


Assuntos
Injúria Renal Aguda/metabolismo , Hemoglobinas/metabolismo , Sepse/metabolismo , Acetaminofen/uso terapêutico , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/prevenção & controle , Anemia Falciforme/metabolismo , Animais , Ponte de Artéria Coronária , Coagulação Intravascular Disseminada/metabolismo , Eriptose , Deformação Eritrocítica , Haptoglobinas/metabolismo , Haptoglobinas/uso terapêutico , Heme/metabolismo , Hemoglobinas/imunologia , Hemólise , Hemopexina/metabolismo , Hemopexina/uso terapêutico , Humanos , Túbulos Renais , Malária/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Sepse/complicações , Sepse/imunologia , Reação Transfusional/metabolismo
20.
Biochim Biophys Acta ; 1784(10): 1448-53, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18342640

RESUMO

Progress in developing a blood substitute is aided by new biotechnologies and a better understanding of the circulatory system. For Hb based solutions, there is still a debate over the best set of fundamental parameters concerning the oxygen affinity which is correlated with the oxidation rate, the cooperativity, the transporter size, and of course the final source of material. Genetic engineering methods have helped discover novel globins, but not yet the quantity necessary for the high demand of blood transfusions. The expanding database of globin properties has indicated that certain individual parameters are coupled, such as the oxygen affinity and the oxidation rate, indicating that one must accept a compromise of the best parameters. After a general introduction of these basic criteria, we will focus on two strategies concerning the size of the oxygen transporter: Hb octamers, and Hb integrated within a nanoparticle.


Assuntos
Substitutos Sanguíneos/uso terapêutico , Hemoglobinas/uso terapêutico , Nanopartículas/uso terapêutico , Transporte Biológico , Substitutos Sanguíneos/síntese química , Substitutos Sanguíneos/química , Engenharia Genética/métodos , Vetores Genéticos , Globinas/química , Globinas/genética , Globinas/uso terapêutico , Haptoglobinas/química , Haptoglobinas/genética , Haptoglobinas/uso terapêutico , Hemoglobinas/química , Hemoglobinas/genética , Humanos , Cinética , Preparações Farmacêuticas/administração & dosagem , Polissacarídeos/química , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA