Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Chembiochem ; 20(15): 1914-1918, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30973186

RESUMO

The biological activity of the glycoprotein hormone erythropoietin (EPO) is dependent mainly on the structure of its N-linked glycans. We aimed to readily attach defined N-glycans to EPO through copper-catalyzed azide alkyne cycloaddition. EPO variants with an alkyne-bearing non-natural amino acid (Plk) at the N-glycosylation sites 24, 38, and 83 were obtained by amber suppression followed by protein purification and refolding. Click conjugation of the alkynyl EPOs with biantennary N-glycan azides provided biologically active site-specifically modified EPO glycoconjugates.


Assuntos
Eritropoetina/síntese química , Polissacarídeos/química , Eritropoetina/química , Humanos , Modelos Moleculares , Estrutura Molecular
2.
Curr Opin Nephrol Hypertens ; 27(5): 351-357, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29847322

RESUMO

PURPOSE OF REVIEW: Sotatercept and luspatercept are recombinant soluble activin type-II receptor-IgG-Fc fusion proteins that are tested in clinical trials for the treatment of various types of anemias, including renal anemia. The mechanism of the action of the novel drugs is incompletely understood, but it seems to be based on the inactivation of soluble proteins of the transforming growth factor-ß (TGFß) family. This review considers pros and cons of the clinical use of the drugs in reference to the current therapy with recombinant erythropoiesis-stimulating agents (ESAs). RECENT FINDINGS: One or more activin type-II receptor (ActRII) ligands appear to inhibit erythroid precursors, for example growth and differentiation factor 11. Trapping of these ligands by the recombinant ActRII fusion proteins, sotatercept and luspatercept increases red blood cell numbers and hemoglobin levels in humans. Reportedly, the novel compounds were well tolerated in trials on healthy volunteers and patients suffering from anemia due to chronic kidney disease or malignancies. On approval, the drugs may prove particularly useful in patients suffering from ineffective erythropoiesis, such as in myelodysplastic syndrome, multiple myeloma or ß-thalassemia, where ESAs are of little use. Independent of their effect on erythropoiesis, ActRII ligand traps were found to exert beneficial effects on renal tissue in experimental animals. SUMMARY: ESAs are likely to remain standard of care in renal anemia. There is a need for a better understanding of the effects of ActRII ligand traps on TGFß-like proteins. The novel drugs have not been approved for sale as therapeutics so far. Their long-term efficacy and safety still needs to be proven, particularly with respect to immunogenicity. Antifibrotic effects may be worthy to be investigated in humans.


Assuntos
Ativinas/farmacologia , Anemia/tratamento farmacológico , Eritropoese/efeitos dos fármacos , Hematínicos/uso terapêutico , Fragmentos Fc das Imunoglobulinas/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Insuficiência Renal Crônica/complicações , Receptores de Ativinas/efeitos dos fármacos , Receptores de Activinas Tipo II , Ativinas/metabolismo , Ativinas/uso terapêutico , Anemia/etiologia , Animais , Eritropoese/fisiologia , Hematínicos/farmacologia , Humanos , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Ligantes , Proteínas Recombinantes de Fusão/uso terapêutico , Fator de Crescimento Transformador beta/antagonistas & inibidores
3.
Arch Biochem Biophys ; 613: 23-30, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27823907

RESUMO

In women breast cancer is still the most commonly diagnosed cancer. This type of cancer is classified as a hormone-dependent tumor. Estrogen receptor (ER) expression and functional status contribute to breast cancer development and progression. Another important factor associated with cancer is hypoxia which is defined as the state of reduced oxygen availability in tissues. Intratumoral hypoxia results in the activation of the hypoxia inducible factors (HIFs). HIFs are heterodimeric transcription factors involved in the regulation of many cellular processes, such as angiogenesis, anaerobic metabolism, cell proliferation/survival, and promotion of metastasis. In this study we evaluated the interplay between hypoxia, HIF stabilization and the ER-α/ß-ratio in several ER-positive breast cancer cell lines. Hypoxia was shown to inhibit ER expression in ER-positive breast cancer cells. Further experiments using the hypoxia mimetic CoCl2 and HIF-1α knockdown cells indicated that the influence of hypoxia on breast cancer cells involves other pathways than the molecular oxygen sensing pathway. Moreover, we demonstrated that MCF-7 cells in long-term culture lost part of their ability to respond to hypoxic incubation. Understanding the relationships between HIF, ER-α and ER-ß expression holds the promise of the development of new therapeutic agents and may provide future advances in prognosis.


Assuntos
Neoplasias da Mama/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Cobalto/química , Progressão da Doença , Estrogênios/metabolismo , Feminino , Humanos , Hipóxia , Células MCF-7 , Oxigênio/química
4.
Cell Physiol Biochem ; 37(6): 2257-64, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26624507

RESUMO

BACKGROUND: Increased red blood cell count (Erythrocytosis) is an important paraneoplastic syndrome of hepatocellular carcinoma (HCC) and is a significant risk factor for lethal lung artery thromboembolism. HCC-associated erythrocytosis is partially caused by the ability of several HCC cells to produce erythropoietin (EPO). Prolyl-4-hydroxylase 2 (PHD2) is an enzyme encoded by the gene EGLN1. The best-known function of PHD2 is to mediate the oxygen-dependent degradation of the labile α-subunit of hypoxia-inducible factor (HIF). However, there is increasing evidence that PHD2 also regulates HIF-independent pathways by interacting with other substrates. METHODS: In the EPO-producing human HCC cell line HepG2, the expression of PHD2 gene was silenced with siRNA. EPO production was estimated using quantitative PCR and ELISA. RESULTS: In HepG2 cells, PHD2 suppresses the activity of TGF-ß1 pathway and consequently maintains the expression of hepatocyte nuclear factor-4α (HNF-4α), an important transcription factor promoting the EPO expression in hepatocytes. PHD2 knockdown caused a marked reduction of EPO production. HIF seemed not to be involved in this biology. CONCLUSION: Our findings show that PHD2 represents a potential contributing factor for HCC-associated erythrocytosis. Selective inhibition of PHD2 in HCC cells might be considered as a new way to manage erythrocytosis in HCC patients.


Assuntos
Carcinoma Hepatocelular/complicações , Eritropoetina/biossíntese , Fator 4 Nuclear de Hepatócito/metabolismo , Neoplasias Hepáticas/complicações , Policitemia/etiologia , Prolil Hidroxilases/metabolismo , Carcinoma Hepatocelular/metabolismo , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Policitemia/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo
5.
Nephrol Dial Transplant ; 30(4): 553-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24748667

RESUMO

Recombinant human erythropoietin (rhEpo, epoetin) has proved beneficial in preventing transfusion-dependent anaemia in patients with chronic kidney disease. Apart from copied epoetins distributed in less regulated markets, 'biosimilar' epoetins have gained currency in many regions, where they compete with the originals and with rhEpo analogues with prolonged survival in circulation ('biobetter'). Recombinant erythropoiesis stimulating agents are potent and well tolerated. However, their production is costly, and they must be administered by the parenteral route. Hence, other anti-anaemia treatments are being evaluated. Clinical trials are being performed with stabilizers of the hypoxia-inducible transcription factors (HIFs), which increase endogenous Epo production. HIF stabilizers are chemical drugs and they are active on oral administration. However, there is fear that they may promote tumour growth. Epo mimetic peptides have also raised expectations. Yet the prototype peginesatide was recalled after just 1 year of its widespread use in the USA because of serious side-effects including cases of death. Most recently, clinical trials have been initiated with sotatercept, a recombinant soluble activin receptor type 2A IgG-Fc fusion protein. Sotatercept binds distinct members of the transforming growth factor-ß family, thereby preventing the inhibitory action of these factors in erythropoiesis. Taken together, rhEpo and its long-acting recombinant analogues will likely remain mainstay of anti-anaemia therapies in the near future.


Assuntos
Ativinas/uso terapêutico , Anemia/tratamento farmacológico , Medicamentos Biossimilares/uso terapêutico , Eritropoetina/uso terapêutico , Hematínicos/uso terapêutico , Humanos
6.
Ann Hematol ; 93(2): 181-92, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24337485

RESUMO

Testing for the presence of specific cell-surface receptors (such as EGFR or HER2) on tumor cells is an integral part of cancer care in terms of treatment decisions and prognosis. Understanding the strengths and limitations of these tests is important because inaccurate results may occur if procedures designed to prevent false-negative or false-positive outcomes are not employed. This review discusses tests commonly used to identify and characterize cell-surface receptors, such as the erythropoietin receptor (EpoR). First, a summary is provided on the biology of the Epo/EpoR system, describing how EpoR is expressed on erythrocytic progenitors and precursors in the bone marrow where it mediates red blood cell production in response to Epo. Second, studies are described that investigated whether erythropoiesis-stimulating agents could stimulate tumor progression in cancer patients and whether EpoR is expressed and functional on tumor cells or on endothelial cells. The methods used in these studies included immunohistochemistry, Northern blotting, Western blotting, and binding assays. This review summarizes the strengths and limitations of these methods. Critically analyzing data from tests for cell-surface receptors such as EpoR requires understanding the techniques utilized and demonstrating that results are consistent with current knowledge about receptor biology.


Assuntos
Northern Blotting/métodos , Western Blotting/métodos , Medula Óssea/metabolismo , Células Eritroides/metabolismo , Imuno-Histoquímica/métodos , Receptores da Eritropoetina/biossíntese , Animais , Células Eritroides/citologia , Regulação da Expressão Gênica/fisiologia , Humanos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores da Eritropoetina/genética
7.
Cell Physiol Biochem ; 32(4): 849-58, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24081025

RESUMO

BACKGROUND: The Aryl Hydrocarbon Receptor Nuclear Translocator (ARNT, HIF-1ß) is a member of the basic-Helix-Loop-Helix PER/ARNT/SIM (bHLH/PAS) protein family and a vital transcriptional regulator regarding development and physiological adaptation processes. ARNT is discussed to be linked with cancer, and other diseases. ARNT is known to be translocated into the cell nucleus, where accumulation of the protein takes place. ARNT is a heterodimerisation partner of the xenobiotic ligand activated Aryl Hydrocarbon Receptor (AhR), the Single Minded proteins (SIM), the cardiovascular helix-loop-helix factor 1 and the Hypoxia Inducible Factor proteins (HIF-α). ARNT is obligatory for HIF-1, HIF-2 and HIF-3 binding to DNA. Whereas degradation of the HIF-α subunits is suppressed by hypoxia, ARNT is generally regarded as constitutively expressed in excess within the cell, and stabilisation is commonly thought to be oxygen-independent. However, we provide evidence that the regulation of ARNT is far more complex. The aim of our study was to reevaluate the regulation of ARNT expression. METHODS: We examined cell lines of different origin like MCF-7 and T47D (human breast cancer), HeLa (human cervix carcinoma), Hep3B and HepG2 (human hepatoma), Kelly (human neuroblastoma), REPC (human kidney) and Cos7 (primary primate kidney) cells. We used immunoblot analysis, densitometry, RT-PCR and transient transfection. RESULTS AND CONCLUSION: Our results show that ARNT protein levels are influenced by hypoxia and hypoxia mimetics such as cobalt(II)-chloride (CoCl2) and dimethyloxalylglycine (DMOG) in a cell line specific manner. We demonstrate that this effect might be triggered by HIF-1α which plays an important role in the process of stabilizing ARNT in hypoxia.


Assuntos
Translocador Nuclear Receptor Aril Hidrocarboneto/metabolismo , Hipóxia Celular/fisiologia , Aminoácidos Dicarboxílicos/farmacologia , Antimutagênicos/farmacologia , Translocador Nuclear Receptor Aril Hidrocarboneto/química , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cobalto/farmacologia , Células HeLa , Humanos , Immunoblotting
8.
Blood ; 118(9): 2395-404, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21652677

RESUMO

Hemoglobin mass is a key factor for maximal exercise capacity. Some athletes apply prohibited techniques and substances with intent to increase hemoglobin mass and physical performance, and this is often difficult to prove directly. Autologous red blood cell transfusion cannot be traced on reinfusion, and also recombinant erythropoietic proteins are detectable only within a certain timeframe. Novel erythropoietic substances, such as mimetics of erythropoietin (Epo) and activators of the Epo gene, may soon enter the sports scene. In addition, Epo gene transfer maneuvers are imaginable. Effective since December 2009, the World Anti-Doping Agency has therefore implemented "Athlete Biologic Passport Operating Guidelines," which are based on the monitoring of several parameters for mature red blood cells and reticulocytes. Blood doping may be assumed, when these parameters change in a nonphysiologic way. Hematologists should be familiar with blood doping practices as they may play an important role in evaluating blood profiles of athletes with respect to manipulations, as contrasted with the established diagnosis of clinical disorders and genetic variations.


Assuntos
Biomarcadores/sangue , Dopagem Esportivo , Detecção do Abuso de Substâncias/normas , Animais , Desempenho Atlético , Melhoramento Biomédico , Contagem de Células Sanguíneas , Dopagem Esportivo/legislação & jurisprudência , Dopagem Esportivo/prevenção & controle , Transfusão de Eritrócitos , Eritropoetina/administração & dosagem , Eritropoetina/biossíntese , Eritropoetina/sangue , Eritropoetina/genética , Eritropoetina/farmacologia , Técnicas de Transferência de Genes , Guias como Assunto , Hematínicos/administração & dosagem , Hematínicos/sangue , Hematínicos/farmacologia , Hematócrito , Hemoglobinas/análise , Humanos , Agências Internacionais/normas , Oxigênio/sangue , Peptídeos Cíclicos/administração & dosagem , Peptídeos Cíclicos/sangue , Peptídeos Cíclicos/farmacologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/sangue , Proteínas Recombinantes/farmacologia , Detecção do Abuso de Substâncias/métodos , Regulação para Cima/efeitos dos fármacos
9.
Transfus Med Hemother ; 40(5): 302-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24273483

RESUMO

Human erythropoietin (Epo) is a 30.4 kDa glycoprotein hormone composed of a single 165 amino acid residues chain to which four glycans are attached. The kidneys are the primary sources of Epo, its synthesis is controlled by hypoxia-inducible transcription factors (HIFs). Epo is an essential factor for the viability and proliferation of erythrocytic progenitors. Whether Epo exerts cytoprotection outside the bone marrow still needs to be clarified. Epo deficiency is the primary cause of the anemia in chronic kidney disease (CKD). Treatment with recombinant human Epo (rhEpo, epoetin) can be beneficial not only in CKD but also for other indications, primarily anemia in cancer patients receiving chemotherapy. Considering unwanted events, the administration of rhEpo or its analogs may increase the incidence of thromboembolism. The expiry of the patents for the original epoetins has initiated the production of similar biological medicinal products ('biosimilars'). Furthermore, analogs (darbepoetin alfa, methoxy PEG-epoetin beta) with prolonged survival in circulation have been developed ('biobetter'). New erythropoiesis-stimulating agents are in clinical trials. These include compounds that augment erythropoiesis directly (e.g. Epo mimetic peptides or activin A binding protein) and chemicals that act indirectly by stimulating endogenous Epo synthesis (HIF stabilizers).

10.
Transfus Med Hemother ; 40(5): 310-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24273484

RESUMO

Anemia is common in intensive care unit (ICU) patients. Red blood cell (RBC) transfusions are mainstays of their treatment and can be life-saving. Allogeneic blood components inherently bear risks of infection and immune reactions. Although these risks are rare in developed countries, recombinant human erythropoietin (rhEpo) and other erythropoiesis-stimulating agents (ESAs) have been considered alternative anti-anemia treatment options. As summarized herein, however, most of the clinical studies suggest that ESAs are not usually advisable in ICU patients unless approved indications exist (e.g., renal disease). First, ESAs act in a delayed way, inducing an increase in reticulocytes only after a lag of 3-4 days. Second, many critically ill patients present with ESA resistance as inflammatory mediators impair erythropoietic cell proliferation and iron availability. Third, the ESA doses used for treatment of ICU patients are very high. Fourth, ESAs are not legally approved for general use in ICU patients. Solely in distinct cases, such as Jehovah's Witnesses who refuse allogeneic blood transfusions due to religious beliefs, ESAs may be considered an exceptional therapy.

11.
Kidney Int ; 81(8): 727-32, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22336988

RESUMO

Antibody-mediated pure red cell aplasia is a very rare but devastating condition affecting patients receiving treatment with erythropoiesis-stimulating agents. New cases continue to emerge, generally in clusters, consistent with an 'environmental' trigger to its pathogenesis. Defining the causes of antibody-mediated pure red cell aplasia is clearly of importance for patients with chronic kidney disease, but any developments in this area may also have relevance to other disease areas as therapeutic delivery of endogenous proteins rapidly increases. This review focuses on the current knowledge regarding the etiology of antibody-mediated pure red cell aplasia and the current approach to therapy.


Assuntos
Hematínicos/efeitos adversos , Hematínicos/imunologia , Aplasia Pura de Série Vermelha/etiologia , Aplasia Pura de Série Vermelha/imunologia , Insuficiência Renal Crônica/tratamento farmacológico , Química Farmacêutica , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Epoetina alfa , Eritropoetina/administração & dosagem , Eritropoetina/efeitos adversos , Eritropoetina/química , Eritropoetina/imunologia , Hematínicos/administração & dosagem , Hematínicos/química , Humanos , Tolerância Imunológica , Multimerização Proteica/imunologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Insuficiência Renal Crônica/diagnóstico , Insuficiência Renal Crônica/terapia , Fatores de Tempo
12.
Front Pharmacol ; 13: 812888, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35847006

RESUMO

Burns are leading causes of mortality and morbidity, including prolonged hospitalization, disfigurement, and disability. Erythropoietin (EPO) is a well-known hormone causing erythropoiesis. However, EPO may play a role in healing acute and chronic wounds due to its anti-inflammatory and pro-regenerative effects. Therefore, the large, prospective, placebo-controlled, randomized, double-blind, multi-center clinical trial "EPO in Burns" was initiated to investigate the effects of EPO versus placebo treatment in severely burned patients. The primary endpoint of "EPO in Burns" was defined as the time elapsed until complete re-epithelialization of a defined split skin graft donor site. Additional analyses of post hoc defined subgroups were performed in view of the primary endpoint. The verum (n 45) and control (n 39) groups were compared with regard to the time it took for study wounds (a predefined split skin graft donor site) to reach the three stages of wound healing (re-epithelialization levels). In addition, the effects of gender (females n 18) and concomitant medications insulin (n 36), non-steroidal anti-inflammatory drugs (NSAIDs) (n 41), and vasopressor agents (n 43) were tested. Life tables were used to compare study groups (EPO vs. placebo) within subgroups. The Cox regression model was applied to evaluate interactions between the study drug (EPO) and concomitant medications for each re-epithelialization level. Using our post hoc defined subgroups, we observed a lower chance of wound healing for women compared to men (in terms of hazard ratio: hr100%: 5.984 [95%-CI: (0.805-44.490), p = 0.080]) in our study population, regardless of the study medication. In addition, results indicated an earlier onset of re-epithelialization in the first days of EPO treatment (EPO: 10% vs. Placebo: 3%). Moreover, the interpretation of the hazard ratio suggested EPO might have a positive, synergistic effect on early stages of re-epithelialization when combined with insulin [hr50%: 1.307 (p = 0.568); hr75%: 1,199 (p = 0.715)], as well as a stabilizing effect on critically ill patients [reduced need for vasopressors in the EPO group (EPO: 44% vs. Placebo 59%)]. However, additional high-quality data from clinical trials designed to address these endpoints are required to gain further insight into these effects.

13.
J Physiol ; 589(Pt 6): 1251-8, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21078592

RESUMO

The hormone erythropoietin (Epo) maintains red blood cell mass by promoting the survival, proliferation and differentiation of erythrocytic progenitors. Circulating Epo originates mainly from fibroblasts in the renal cortex. Epo production is controlled at the transcriptional level. Hypoxia attenuates the inhibition of the Epo promoter by GATA-2. More importantly, hypoxia promotes the availability of heterodimeric (α/ß) hypoxia-inducible transcription factors (predominantly HIF-2) which stimulate the Epo enhancer. The HIFs are inactivated in normoxia by enzymatic hydroxylation of their α-subunits. Three HIF-α prolyl hydroxylases (PHD-1, -2 and -3) initiate proteasomal degradation of HIF-α, while an asparaginyl hydroxylase ('factor inhibiting HIF-1', FIH-1) inhibits the transactivation potential. The HIF-α hydroxylases contain Fe(2+) and require 2-oxoglutarate as co-factor. The in vivo response is dynamic, i.e. the concentration of circulating Epo increases initially greatly following an anaemic or hypoxaemic stimulus and then declines despite continued hypoxia. Epo and angiotensin II collaborate in the maintenance of the blood volume. Whether extra-renal sites (brain, skin) modulate renal Epo production is a matter of debate. Epo overproduction results in erythrocytosis. Epo deficiency is the primary cause of the anaemia in chronic kidney disease and a contributing factor in the anaemias of chronic inflammation and cancer. Here, recombinant analogues can substitute for the hormone.


Assuntos
Eritropoese/fisiologia , Eritropoetina/biossíntese , Hipóxia/metabolismo , Rim/metabolismo , Angiotensina II/metabolismo , Animais , Eritrócitos/metabolismo , Eritropoetina/fisiologia , Retroalimentação Fisiológica/fisiologia , Humanos
14.
Cell Physiol Biochem ; 28(5): 805-12, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22178933

RESUMO

BACKGROUND: Radiotherapy is one of the most widely used treatments for cancer. The benefit of radiation is known to be negatively affected by tumor hypoxia and the expression of hypoxia-inducible factors (HIF), respectively. HIF-1α/ ß and HIF-2α/ ß are transcriptional activators of oxygen-regulated genes. The aim of the study was to examine cell type-specific effects of HIF-1α and -2α knockdown or oxygen-independent HIF-stabilisation on radiosensitivity. METHODS: Herein, we treated four different wildtype and HIF-1α- or HIF-2α-deficient human cancer cell lines, cultured under normoxic or hypoxic conditions, with ionising radiation in doses from 2 to 6 Gy and examined clonogenic survival. Furthermore, the cells were partly preincubated with a HIF-stabiliser (di-tert-butyroyl-oxymethyl-2,4-pyridine-dicarboxylate, (t)Bu-2,4-PDC). RESULTS: The results show that both hypoxia exposure and treatment with (t)Bu-2,4-PDC increased the radioresistance of human cancer cells. The HIF-mediated decrease of radioresponsiveness induced by the chemical stabiliser emerged to be as strong as the one caused by hypoxia. Clonogenic survival assays furthermore revealed that HIF-1 expression enhanced resistance to radiation, whereas knocking-down HIF-1 increased the sensitivity to radiation under normoxic as well as under hypoxic conditions. CONCLUSION: These data extend previous observations of HIF-1α and broaden the view by showing HIF-2α inverse correlation between HIF expression and prognosis for the outcome of radiotherapy.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Raios gama , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Tolerância a Radiação/efeitos dos fármacos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Hipóxia Celular , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias/radioterapia , Ácidos Pipecólicos/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo
15.
Strahlenther Onkol ; 187(7): 393-400, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21713389

RESUMO

BACKGROUND: Curcumin, a commonly used spice, affects the activities of cytokines, enzymes, and transcription factors that are linked to inflammation. Furthermore, curcumin has been assigned tumor growth inhibiting effects, possibly mediated by promoting hypoxia-inducible factor (HIF) degradation. HIFs are transcription factors that play a central role in the adaptation and response to low oxygen levels in metazoan cells. However, curcumin also exhibits properties of an iron chelator indicating its potential of inhibiting HIF-α prolyl hydroxylase (PHD) activity. METHODS: We were interested in clarifying these divergent actions of curcumin in due consideration of the effects on radio-therapy. Thus, concentration- and time-dependent effects of curcumin on HIF-α and -ß protein levels and activity in hepatoma and breast carcinoma cell cultures under normoxic and hypoxic conditions were studied. RESULTS: It was shown that HIF-1α accumulated in normoxia after the application of higher doses of the drug. Curcumin proved to lower HIF-1α and HIF-2α protein levels in hypoxia. HIF-1ß (ARNT; arylhydrocarbon nuclear translocator) protein levels and HIF transcriptional activity were reduced in normoxia and hypoxia after 4 h and 24 h incubation periods. Furthermore, curcumin treatment negatively impacted on clonogenic cell survival of Hep3B hepatoma and MCF-7 breast carcinoma cells. CONCLUSION: Effects of curcumin on cell growth and survival factor expression suggest its potential benefit in the treatment of cancer without a direct radiosensitizing influence of curcumin on these cells.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Cuminum , Neoplasias Hepáticas/patologia , Fitoterapia , Preparações de Plantas/farmacologia , Radiossensibilizantes/farmacologia , Apoptose/efeitos da radiação , Translocador Nuclear Receptor Aril Hidrocarboneto/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Divisão Celular/efeitos da radiação , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Masculino , Ensaio Tumoral de Célula-Tronco
16.
Bioessays ; 31(3): 344-8, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19260015

RESUMO

Erythropoietin (EPO), long appreciated as the chief endocrine regulator of red blood cell formation, is now recognized to exert many additional functions outside the bone marrow. Thus, the quest is on to define the full range of EPO functions in the physiology and pathology of non-hematopoietic tissues. Two recent studies in man and mice have highlighted the importance of the mammalian skin as one peripheral tissue with a previously unsuspected role in EPO biology; both, as a target and as a source of EPO. In addition, the skin has been proposed to function as an oxygen sensor. The present hypothesis essay critically reviews the currently available evidence for this and provides a unifying theoretical scenario for intracutaneous EPO functions and for a potential role of the skin in the control of EPO production. Mainly, we propose that the skin itself directly contributes to the up-regulation of EPO plasma levels in response to hypoxia.


Assuntos
Eritropoetina/metabolismo , Oxigênio/metabolismo , Pele/metabolismo , Animais , Humanos , Hipóxia/metabolismo
17.
Exp Dermatol ; 19(1): 65-7, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19645852

RESUMO

Erythropoietin (EPO) is now appreciated for not only drive erythopoiesis, but also to exert additional functions. Since we had previously shown that human hair follicles (HFs) are both an extra-renal source and an extra-medullary target of EPO, we have now studied whether one such function is the regulation of HF pigmentation. Human anagen VI HFs were treated with EPO (100 IU/ml) in serum-free organ culture. Unexpectedly, we noticed greatly divergent pigmentary effects of EPO, since both up- and down-regulation of HF melanin content and tyrosinase activity in situ was seen in HF derived from different individuals. These divergent effects could not be attributed to differences in skin regions, the total HF melanocyte number or specific traits of individual HF donors. Our pilot study provides first evidence suggesting that EPO may modulate normal human melanocyte functions under physiologically relevant conditions in situ.


Assuntos
Eritropoetina/metabolismo , Folículo Piloso/metabolismo , Melanócitos/metabolismo , Humanos , Técnicas de Cultura de Órgãos , Projetos Piloto
18.
Am J Hematol ; 85(10): 771-80, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20706990

RESUMO

After the patents of biopharmaceuticals have expired, based on specific regulatory approval pathways copied products ("biosimilars" or "follow-on biologics") have been launched in the EU. This article summarizes experiences with hematopoietic medicines, namely the epoetins (two biosimilars traded under five different brand names) and the filgrastims (two biosimilars, six brand names). Physicians and pharmacists should be familiar with the legal and pharmacological specialities of biosimilars: The production process can differ from that of the original, clinical indications can be extrapolated, glycoproteins contain varying isoforms, the formulation may differ from the original, and biopharmaceuticals are potentially immunogenic. Only on proof of quality, efficacy and safety, biosimilars are a viable option because of their lower costs.


Assuntos
Produtos Biológicos/uso terapêutico , Eritropoetina/uso terapêutico , Anemia/etiologia , Anemia/terapia , Antineoplásicos/efeitos adversos , Produtos Biológicos/economia , Produtos Biológicos/farmacocinética , Produtos Biológicos/normas , Doença Crônica , Uso de Medicamentos , Eritropoetina/economia , Eritropoetina/imunologia , Eritropoetina/farmacocinética , União Europeia , Filgrastim , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Guias como Assunto , Humanos , Isoanticorpos/biossíntese , Nefropatias/sangue , Nefropatias/complicações , Legislação de Medicamentos , Marketing , Neoplasias/sangue , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Patentes como Assunto , Polietilenoglicóis , Controle de Qualidade , Proteínas Recombinantes/economia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapêutico , Aplasia Pura de Série Vermelha/etiologia , Equivalência Terapêutica , Organização Mundial da Saúde
19.
Ann Hematol ; 88(5): 411-5, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19031076

RESUMO

Anemia is a common and serious complication of malaria due to Plasmodium falciparum infection, a major health problem in tropical areas. Herein, the relation was investigated between the levels of circulating erythropoietin (EPO) and immunomodulatory cytokines in response to chloroquine treatment. Thirty-seven healthy control subjects and 40 patients with acute P. falciparum infection were included in the study. All subjects were adult male Sudanese. Blood samples were collected before chloroquine administration (25 mg/kg body weight, orally on three consecutive days) and 3 and 30 days after start of the therapy. Measurements included routine hematological parameters and the concentrations of immunoreactive EPO, tumor necrosis factor-alpha (TNF-alpha), interleukin 1alpha (IL-1), IL-6, and interferon gamma (INF-gamma). Chloroquine treatment led to a decrease in EPO levels in the control subjects but an increase in malaria patients at day 30. The latter was likely due to the anti-inflammatory action of the drug because INF-gamma, IL-1, and IL-6 concentrations declined on chloroquine treatment. Based on these findings, we propose that an impaired EPO production in association with a prolonged elevation of certain inflammatory cytokines can contribute to the anemia in some malaria patients which can be reversed by chloroquine therapy.


Assuntos
Cloroquina/administração & dosagem , Citocinas/efeitos dos fármacos , Eritropoetina/sangue , Malária Falciparum/tratamento farmacológico , Adulto , Estudos de Casos e Controles , Citocinas/sangue , Humanos , Fatores Imunológicos/sangue , Interferon gama/sangue , Interleucina-1/sangue , Interleucina-6/sangue , Malária Falciparum/sangue , Masculino , Sudão
20.
Eur J Appl Physiol ; 106(4): 599-604, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19363619

RESUMO

Oxidative stress occurs at altitude, and physical exertion might enhance this stress. In the present study, we investigated the combined effects of exercise and moderate altitude on redox balance in ten endurance exercising biathletes, and five sedentary volunteers during a 6-week-stay at 2,800 m. As a marker for oxidative stress, hydrogen peroxide (H(2)O(2)) was analyzed by the biosensor measuring system Ecocheck, and 8-iso prostaglandin F2alpha (8-iso PGF2alpha) was determined by enzyme immunoassay in exhaled breath condensate (EBC). To determine the whole blood antioxidative capacity, we measured reduced glutathione (GSH) enzymatically using Ellman's reagent. Exercising athletes and sedentary volunteers showed increased levels of oxidative markers at moderate altitude, contrary to our expectations; there was no difference between both groups. Therefore, all subjects' data were pooled to examine the oxidative stress response exclusively due to altitude exposure. H(2)O(2) levels increased at altitude and remained elevated for 3 days after returning to sea level (p < or = 0.05). On the other hand, 8-iso PGF2alpha levels showed a tendency to increase at altitude, but declined immediately after returning to sea level (p < or = 0.001). Hypoxic exposure during the first day at altitude resulted in elevated GSH levels (p < or = 0.05), that decreased during prolonged sojourn at altitude (p < or = 0.001). In conclusion, a stay at moderate altitude for up to 6 weeks increases markers of oxidative stress in EBC independent of additional endurance training. Notably, this oxidative stress is still detectable 3 days upon return to sea level.


Assuntos
Altitude , Testes Respiratórios , Expiração/fisiologia , Estresse Oxidativo/fisiologia , Resistência Física/fisiologia , Esforço Físico/fisiologia , Aptidão Física/fisiologia , Espécies Reativas de Oxigênio/análise , Feminino , Humanos , Masculino , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA