Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
PLoS Pathog ; 18(2): e1009977, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35192672

RESUMEN

Plasmodium falciparum exports ~10% of its proteome into its host erythrocyte to modify the host cell's physiology. The Plasmodium export element (PEXEL) motif contained within the N-terminus of most exported proteins directs the trafficking of those proteins into the erythrocyte. To reach the host cell, the PEXEL motif of exported proteins is processed by the endoplasmic reticulum (ER) resident aspartyl protease plasmepsin V. Then, following secretion into the parasite-encasing parasitophorous vacuole, the mature exported protein must be unfolded and translocated across the parasitophorous vacuole membrane by the Plasmodium translocon of exported proteins (PTEX). PTEX is a protein-conducting channel consisting of the pore-forming protein EXP2, the protein unfoldase HSP101, and structural component PTEX150. The mechanism of how exported proteins are specifically trafficked from the parasite's ER following PEXEL cleavage to PTEX complexes on the parasitophorous vacuole membrane is currently not understood. Here, we present evidence that EXP2 and PTEX150 form a stable subcomplex that facilitates HSP101 docking. We also demonstrate that HSP101 localises both within the parasitophorous vacuole and within the parasite's ER throughout the ring and trophozoite stage of the parasite, coinciding with the timeframe of protein export. Interestingly, we found that HSP101 can form specific interactions with model PEXEL proteins in the parasite's ER, irrespective of their PEXEL processing status. Collectively, our data suggest that HSP101 recognises and chaperones PEXEL proteins from the ER to the parasitophorous vacuole and given HSP101's specificity for the EXP2-PTEX150 subcomplex, this provides a mechanism for how exported proteins are specifically targeted to PTEX for translocation into the erythrocyte.


Asunto(s)
Parásitos , Plasmodium falciparum , Animales , Eritrocitos/parasitología , Parásitos/metabolismo , Plasmodium falciparum/metabolismo , Transporte de Proteínas/fisiología , Proteínas Protozoarias/metabolismo
3.
Artículo en Inglés | MEDLINE | ID: mdl-33014890

RESUMEN

An important component in host resistance to malaria infection are inherited mutations that give rise to abnormalities and deficiencies in erythrocyte proteins and enzymes. Understanding how such mutations confer protection against the disease may be useful for developing new treatment strategies. A mouse ENU-induced mutagenesis screen for novel malaria resistance-conferring mutations identified a novel non-sense mutation in the gene encoding porphobilinogen deaminase (PBGD) in mice, denoted here as PbgdMRI58155. Heterozygote PbgdMRI58155 mice exhibited ~50% reduction in cellular PBGD activity in both mature erythrocytes and reticulocytes, although enzyme activity was ~10 times higher in reticulocytes than erythrocytes. When challenged with blood-stage P. chabaudi, which preferentially infects erythrocytes, heterozygote mice showed a modest but significant resistance to infection, including reduced parasite growth. A series of assays conducted to investigate the mechanism of resistance indicated that mutant erythrocyte invasion by P. chabaudi was normal, but that following intraerythrocytic establishment a significantly greater proportions of parasites died and therefore, affected their ability to propagate. The Plasmodium resistance phenotype was not recapitulated in Pbgd-deficient mice infected with P. berghei, which prefers reticulocytes, or when P. falciparum was cultured in erythrocytes from patients with acute intermittent porphyria (AIP), which had modest (20-50%) reduced levels of PBGD. Furthermore, the growth of Pbgd-null P. falciparum and Pbgd-null P. berghei parasites, which grew at the same rate as their wild-type counterparts in normal cells, were not affected by the PBGD-deficient background of the AIP erythrocytes or Pbgd-deficient mice. Our results confirm the dispensability of parasite PBGD for P. berghei infection and intraerythrocytic growth of P. falciparum, but for the first time identify a requirement for host erythrocyte PBGD by P. chabaudi during in vivo blood stage infection.


Asunto(s)
Malaria , Plasmodium chabaudi , Porfiria Intermitente Aguda , Animales , Eritrocitos , Humanos , Ratones , Plasmodium berghei/genética , Plasmodium falciparum
4.
mBio ; 10(3)2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31164473

RESUMEN

Plasmodium parasites must export proteins into their erythrocytic host to survive. Exported proteins must cross the parasite plasma membrane (PPM) and the parasitophorous vacuolar membrane (PVM) encasing the parasite to access the host cell. Crossing the PVM requires protein unfolding and passage through a translocon, the Plasmodium translocon of exported proteins (PTEX). In this study, we provide the first direct evidence that heat shock protein 101 (HSP101), a core component of PTEX, unfolds proteins for translocation across the PVM by creating transgenic Plasmodium parasites in which the unfoldase and translocation functions of HSP101 have become uncoupled. Strikingly, while these parasites could export native proteins, they were unable to translocate soluble, tightly folded reporter proteins bearing the Plasmodium export element (PEXEL) across the PVM into host erythrocytes under the same conditions. In contrast, an identical PEXEL reporter protein but harboring a transmembrane domain could be exported, suggesting that a prior unfolding step occurs at the PPM. Together, these results demonstrate that the export of parasite proteins is dependent on how these proteins are presented to the secretory pathway before they reach PTEX as well as their folded status. Accordingly, only tightly folded soluble proteins secreted into the vacuolar space and not proteins containing transmembrane domains or the majority of erythrocyte-stage exported proteins have an absolute requirement for the full unfoldase activity of HSP101 to be exported.IMPORTANCE The Plasmodium parasites that cause malaria export hundreds of proteins into their host red blood cell (RBC). These exported proteins drastically alter the structural and functional properties of the RBC and play critical roles in parasite virulence and survival. To access the RBC cytoplasm, parasite proteins must pass through the Plasmodium translocon of exported proteins (PTEX) located at the membrane interfacing the parasite and host cell. Our data provide evidence that HSP101, a component of PTEX, serves to unfold protein cargo requiring translocation. We also reveal that addition of a transmembrane domain to soluble cargo influences its ability to be translocated by parasites in which the HSP101 motor and unfolding activities have become uncoupled. Therefore, we propose that proteins with transmembrane domains use an alternative unfolding pathway prior to PTEX to facilitate export.


Asunto(s)
Eritrocitos/parasitología , Proteínas de Choque Térmico/metabolismo , Interacciones Huésped-Parásitos , Plasmodium berghei/genética , Desplegamiento Proteico , Proteínas Protozoarias/metabolismo , Animales , Femenino , Proteínas de Choque Térmico/genética , Ratones , Ratones Endogámicos BALB C , Plasmodium berghei/metabolismo , Transporte de Proteínas , Proteínas Protozoarias/genética , Solubilidad
5.
Cell Microbiol ; 21(4): e13009, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30656810

RESUMEN

Plasmodium parasites that cause the disease malaria have developed an elaborate trafficking pathway to facilitate the export of hundreds of effector proteins into their host cell, the erythrocyte. In this review, we outline how certain effector proteins contribute to parasite survival, virulence, and immune evasion. We also highlight how parasite proteins destined for export are recognised at the endoplasmic reticulum to facilitate entry into the export pathway and how the effector proteins are able to transverse the bounding parasitophorous vaculoar membrane via the Plasmodium translocon of exported proteins to gain access to the host cell. Some of the gaps in our understanding of the export pathway are also presented. Finally, we examine the degree of conservation of some of the key components of the Plasmodium export pathway in closely related apicomplexan parasites, which may provide insight into how the diverse apicomplexan parasites have adapted to survival pressures encountered within their respective host cells.


Asunto(s)
Eritrocitos/metabolismo , Eritrocitos/parasitología , Malaria/metabolismo , Malaria/parasitología , Proteínas Protozoarias/metabolismo , Animales , Humanos , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Transporte de Proteínas/fisiología , Proteínas Protozoarias/genética
6.
FEBS J ; 285(11): 2037-2055, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29637707

RESUMEN

The pathogenic nature of malaria infections is due in part to the export of hundreds of effector proteins that actively remodel the host erythrocyte. The Plasmodium translocon of exported proteins (PTEX) has been shown to facilitate the trafficking of proteins into the host cell, a process that is essential for the survival of the parasite. The role of the auxiliary PTEX component PTEX88 remains unclear, as previous attempts to elucidate its function through reverse genetic approaches showed that in contrast to the core components PTEX150 and HSP101, knockdown of PTEX88 did not give rise to an export phenotype. Here, we have used biochemical approaches to understand how PTEX88 assembles within the translocation machinery. Proteomic analysis of the PTEX88 interactome showed that PTEX88 interacts closely with HSP101 but has a weaker affinity with the other core constituents of PTEX. PTEX88 was also found to associate with other PV-resident proteins, including chaperones and members of the exported protein-interacting complex that interacts with the major virulence factor PfEMP1, the latter contributing to cytoadherence and parasite virulence. Despite being expressed for the duration of the blood-stage life cycle, PTEX88 was only discretely observed at the parasitophorous vacuole membrane during ring stages and could not always be detected in the major high molecular weight complex that contains the other core components of PTEX, suggesting that its interaction with the PTEX complex may be dynamic. Together, these data have enabled the generation of an updated model of PTEX that now includes how PTEX88 assembles within the complex.


Asunto(s)
Interacciones Huésped-Parásitos/genética , Malaria Falciparum/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Animales , Eritrocitos/parasitología , Humanos , Estadios del Ciclo de Vida/genética , Malaria Falciparum/parasitología , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Plasmodium falciparum/patogenicidad , Transporte de Proteínas/genética , Proteómica
7.
Cell Microbiol ; 20(8): e12844, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29582546

RESUMEN

The inner membrane complex and the apical secretory organelles are defining features of apicomplexan parasites. Despite their critical roles, the mechanisms behind the biogenesis of these structures in the malaria parasite Plasmodium falciparum are still poorly defined. We here show that decreasing expression of the P. falciparum homologue of the conserved endolysomal escorter Sortilin-VPS10 prevents the formation of the inner membrane complex and abrogates the generation of new merozoites. Moreover, protein trafficking to the rhoptries, the micronemes, and the dense granules is disrupted, which leads to the accumulation of apical complex proteins in the endoplasmic reticulum and the parasitophorous vacuole. We further show that protein export to the erythrocyte and transport through the constitutive secretory pathway are functional. Taken together, our results suggest that the malaria parasite P. falciparum Sortilin has potentially broader functions than most of its other eukaryotic counterparts.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Merozoítos/crecimiento & desarrollo , Biogénesis de Organelos , Plasmodium falciparum/crecimiento & desarrollo , Proteínas Adaptadoras del Transporte Vesicular/genética , Técnicas de Silenciamiento del Gen , Transporte de Proteínas
8.
Nat Commun ; 8: 16044, 2017 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-28691708

RESUMEN

The malaria parasite, Plasmodium falciparum, displays the P. falciparum erythrocyte membrane protein 1 (PfEMP1) on the surface of infected red blood cells (RBCs). We here examine the physical organization of PfEMP1 trafficking intermediates in infected RBCs and determine interacting partners using an epitope-tagged minimal construct (PfEMP1B). We show that parasitophorous vacuole (PV)-located PfEMP1B interacts with components of the PTEX (Plasmodium Translocon of EXported proteins) as well as a novel protein complex, EPIC (Exported Protein-Interacting Complex). Within the RBC cytoplasm PfEMP1B interacts with components of the Maurer's clefts and the RBC chaperonin complex. We define the EPIC interactome and, using an inducible knockdown approach, show that depletion of one of its components, the parasitophorous vacuolar protein-1 (PV1), results in altered knob morphology, reduced cell rigidity and decreased binding to CD36. Accordingly, we show that deletion of the Plasmodium berghei homologue of PV1 is associated with attenuation of parasite virulence in vivo.


Asunto(s)
Interacciones Huésped-Patógeno , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Proteínas Portadoras/metabolismo , Adhesión Celular , Femenino , Técnicas de Silenciamiento del Gen , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Plasmodium berghei/genética , Plasmodium falciparum/patogenicidad , Transporte de Proteínas
9.
Glob Chang Biol ; 23(11): 4483-4496, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28447373

RESUMEN

Climate change and ocean acidification are altering marine ecosystems and, from a human perspective, creating both winners and losers. Human responses to these changes are complex, but may result in reduced government investments in regulation, resource management, monitoring and enforcement. Moreover, a lack of peoples' experience of climate change may drive some towards attributing the symptoms of climate change to more familiar causes such as management failure. Taken together, we anticipate that management could become weaker and less effective as climate change continues. Using diverse case studies, including the decline of coral reefs, coastal defences from flooding, shifting fish stocks and the emergence of new shipping opportunities in the Arctic, we argue that human interests are better served by increased investments in resource management. But greater government investment in management does not simply mean more of "business-as-usual." Management needs to become more flexible, better at anticipating and responding to surprise, and able to facilitate change where it is desirable. A range of technological, economic, communication and governance solutions exists to help transform management. While not all have been tested, judicious application of the most appropriate solutions should help humanity adapt to novel circumstances and seek opportunity where possible.


Asunto(s)
Cambio Climático , Conservación de los Recursos Naturales , Animales , Arrecifes de Coral , Ecosistema , Peces , Humanos , Motivación , Océanos y Mares
10.
Cell Microbiol ; 19(8)2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28205319

RESUMEN

Plasmodium parasites must invade erythrocytes in order to cause the disease malaria. The invasion process involves the coordinated secretion of parasite proteins from apical organelles that include the rhoptries. The rhoptry is comprised of two compartments: the neck and the bulb. Rhoptry neck proteins are involved in host cell adhesion and formation of the tight junction that forms between the invading parasite and erythrocyte, whereas the role of rhoptry bulb proteins remains ill-defined due to the lack of functional studies. In this study, we show that the rhoptry-associated protein (RAP) complex is not required for rhoptry morphology or erythrocyte invasion. Instead, post-invasion when the parasite is bounded by a parasitophorous vacuolar membrane (PVM), the RAP complex facilitates the survival of the parasite in its new intracellular environment. Consequently, conditional knockdown of members of the RAP complex leads to altered PVM structure, delayed intra-erythrocytic growth, and reduced parasitaemias in infected mice. This study provides evidence that rhoptry bulb proteins localising to the parasite-host cell interface are not simply by-products of the invasion process but contribute to the growth of Plasmodium in vivo.


Asunto(s)
Eritrocitos/parasitología , Interacciones Huésped-Patógeno , Plasmodium berghei/fisiología , Proteínas Protozoarias/metabolismo , Vacuolas/parasitología , Factores de Virulencia/metabolismo , Animales , Modelos Animales de Enfermedad , Malaria/parasitología , Ratones Endogámicos BALB C
11.
Cell Microbiol ; 18(3): 399-412, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26347246

RESUMEN

Export of most malaria proteins into the erythrocyte cytosol requires the Plasmodium translocon of exported proteins (PTEX) and a cleavable Plasmodium export element (PEXEL). In contrast, the contribution of PTEX in the liver stages and export of liver stage proteins is unknown. Here, using the FLP/FRT conditional mutatagenesis system, we generate transgenic Plasmodium berghei parasites deficient in EXP2, the putative pore-forming component of PTEX. Our data reveal that EXP2 is important for parasite growth in the liver and critical for parasite transition to the blood, with parasites impaired in their ability to generate a patent blood-stage infection. Surprisingly, whilst parasites expressing a functional PTEX machinery can efficiently export a PEXEL-bearing GFP reporter into the erythrocyte cytosol during a blood stage infection, this same reporter aggregates in large accumulations within the confines of the parasitophorous vacuole membrane during hepatocyte growth. Notably HSP101, the putative molecular motor of PTEX, could not be detected during the early liver stages of infection, which may explain why direct protein translocation of this soluble PEXEL-bearing reporter or indeed native PEXEL proteins into the hepatocyte cytosol has not been observed. This suggests that PTEX function may not be conserved between the blood and liver stages of malaria infection.


Asunto(s)
Malaria/parasitología , Plasmodium berghei/patogenicidad , Proteínas Protozoarias/metabolismo , Animales , Animales Modificados Genéticamente , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Choque Térmico/metabolismo , Interacciones Huésped-Parásitos , Hígado/parasitología , Ratones , Plasmodium berghei/genética , Transporte de Proteínas/genética , Proteínas Protozoarias/genética , Tetraciclinas/farmacología
12.
Nature ; 511(7511): 587-91, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25043043

RESUMEN

During the blood stages of malaria, several hundred parasite-encoded proteins are exported beyond the double-membrane barrier that separates the parasite from the host cell cytosol. These proteins have a variety of roles that are essential to virulence or parasite growth. There is keen interest in understanding how proteins are exported and whether common machineries are involved in trafficking the different classes of exported proteins. One potential trafficking machine is a protein complex known as the Plasmodium translocon of exported proteins (PTEX). Although PTEX has been linked to the export of one class of exported proteins, there has been no direct evidence for its role and scope in protein translocation. Here we show, through the generation of two parasite lines defective for essential PTEX components (HSP101 or PTEX150), and analysis of a line lacking the non-essential component TRX2 (ref. 12), greatly reduced trafficking of all classes of exported proteins beyond the double membrane barrier enveloping the parasite. This includes proteins containing the PEXEL motif (RxLxE/Q/D) and PEXEL-negative exported proteins (PNEPs). Moreover, the export of proteins destined for expression on the infected erythrocyte surface, including the major virulence factor PfEMP1 in Plasmodium falciparum, was significantly reduced in PTEX knockdown parasites. PTEX function was also essential for blood-stage growth, because even a modest knockdown of PTEX components had a strong effect on the parasite's capacity to complete the erythrocytic cycle both in vitro and in vivo. Hence, as the only known nexus for protein export in Plasmodium parasites, and an essential enzymic machine, PTEX is a prime drug target.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Malaria/parasitología , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Eritrocitos/metabolismo , Eritrocitos/parasitología , Proteínas de Choque Térmico/genética , Humanos , Estadios del Ciclo de Vida/fisiología , Complejos Multiproteicos/metabolismo , Transporte de Proteínas/genética , Proteínas Protozoarias/genética , Vacuolas/metabolismo , Vacuolas/parasitología
13.
Mol Microbiol ; 89(6): 1167-86, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23869529

RESUMEN

Plasmodium parasites remodel their vertebrate host cells by translocating hundreds of proteins across an encasing membrane into the host cell cytosol via a putative export machinery termed PTEX. Previously PTEX150, HSP101 and EXP2 have been shown to be bona fide members of PTEX. Here we validate that PTEX88 and TRX2 are also genuine members of PTEX and provide evidence that expression of PTEX components are also expressed in early gametocytes, mosquito and liver stages, consistent with observations that protein export is not restricted to asexual stages. Although amenable to genetic tagging, HSP101, PTEX150, EXP2 and PTEX88 could not be genetically deleted in Plasmodium berghei, in keeping with the obligatory role this complex is postulated to have in maintaining normal blood-stage growth. In contrast, the putative thioredoxin-like protein TRX2 could be deleted, with knockout parasites displaying reduced grow-rates, both in vivo and in vitro, and reduced capacity to cause severe disease in a cerebral malaria model. Thus, while not essential for parasite survival, TRX2 may help to optimize PTEX activity. Importantly, the generation of TRX2 knockout parasites that display altered phenotypes provides a much-needed tool to dissect PTEX function.


Asunto(s)
Parasitemia/parasitología , Plasmodium berghei/enzimología , Plasmodium berghei/patogenicidad , Tiorredoxinas/metabolismo , Factores de Virulencia/metabolismo , Animales , Modelos Animales de Enfermedad , Eliminación de Gen , Malaria Cerebral/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Plasmodium berghei/genética , Plasmodium berghei/crecimiento & desarrollo , Análisis de Supervivencia , Tiorredoxinas/genética , Virulencia , Factores de Virulencia/genética
14.
PLoS One ; 8(4): e61482, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23658610

RESUMEN

Protein export into the host red blood cell is one of the key processes in the pathobiology of the malaria parasite Plasmodiumtrl falciparum, which extensively remodels the red blood cell to ensure its virulence and survival. In this study, we aimed to shed further light on the protein export mechanisms in the rodent malaria parasite P. berghei and provide further proof of the conserved nature of host cell remodeling in Plasmodium spp. Based on the presence of an export motif (R/KxLxE/Q/D) termed PEXEL (Plasmodium export element), we have generated transgenic P. berghei parasite lines expressing GFP chimera of putatively exported proteins and analysed one of the newly identified exported proteins in detail. This essential protein, termed PbCP1 (P. berghei Cleft-like Protein 1), harbours an atypical PEXEL motif (RxLxY) and is further characterised by two predicted transmembrane domains (2TMD) in the C-terminal end of the protein. We have functionally validated the unusual PEXEL motif in PbCP1 and analysed the role of the 2TMD region, which is required to recruit PbCP1 to discrete membranous structures in the red blood cell cytosol that have a convoluted, vesico-tubular morphology by electron microscopy. Importantly, this study reveals that rodent malaria species also induce modifications to their host red blood cell.


Asunto(s)
Citosol/parasitología , Eritrocitos/parasitología , Plasmodium berghei/metabolismo , Proteínas Protozoarias/química , Secuencia de Aminoácidos , Animales , Citosol/ultraestructura , Eritrocitos/ultraestructura , Expresión Génica , Proteínas Fluorescentes Verdes , Estadios del Ciclo de Vida/genética , Malaria/parasitología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Plasmodium berghei/genética , Plasmodium berghei/ultraestructura , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
15.
Stem Cells Dev ; 15(1): 40-8, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16522161

RESUMEN

CD163, the hemoglobin-haptoglobin receptor, has been reported to be expressed exclusively on monocyte/ macrophages. Here we demonstrate that CD163 is also expressed by a subpopulation of hematopoietic stem/progenitor cells. Flow cytometric analysis shows that 1.9 +/- 1.3% (+/-SD, n = 16) of adult bone marrow and 2.0 +/- 1.8% (n = 8) of umbilical cord blood CD34(+) cells express cell-surface CD163, and 69.1 +/- 16.9% (n = 9) and 79.7 +/- 22.4% (n = 8) of the respective cells contain the CD163 protein intracellularly. The expression of CD163 by CD34(+) cells was confirmed by western blot analysis of cell lysates. Transcripts corresponding to the known predominant and variant 1 forms of CD163 were amplified via RT-PCR from CD34(+) cell-derived mRNA. A new variant (K11) with a deletion at the start of exon 15 was also detected. The deleted region contains a PKCalpha phosphorylation site and an amino acid sequence (YREM) that may support efficient receptor endocytosis. The addition of activating anti-CD163 antibodies increased the growth and differentiation of erythroid progenitors in colony-forming assays. These data suggest that hemoglobin may mediate a stimulatory effect on erythropoiesis through the activation of CD163 on hematopoietic progenitor cells.


Asunto(s)
Antígenos CD/sangre , Antígenos de Diferenciación Mielomonocítica/sangre , Células de la Médula Ósea/fisiología , Células Precursoras Eritroides/fisiología , Células Madre Hematopoyéticas/metabolismo , Receptores de Superficie Celular/sangre , Receptores de Superficie Celular/metabolismo , Empalme Alternativo , Antígenos CD34/sangre , Secuencia de Bases , Diferenciación Celular , Células Cultivadas , Expresión Génica , Humanos , Datos de Secuencia Molecular , Homología de Secuencia , Cordón Umbilical/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...