Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtrer
1.
São Paulo; s.n; s.n; 2023. 111 p. tab, graf, ilus.
Thèse de Portugais | LILACS | ID: biblio-1437605

RÉSUMÉ

O envelhecimento é um processo fisiológico que traz consigo uma série de alterações no organismo que se estendem até o nível molecular. Diante disto, este é um processo complexo que afeta diversos tecidos, sendo um deles o hematopoético, local onde, através de interações da Célula Tronco Hematopoética (CTH) com o ambiente ao seu redor, incluindo a Célula Tronco Mesenquimal (CTM), ocorre a hematopoese. Embora já sejam descritas na literatura algumas alterações na medula óssea consequentes do envelhecimento, os mecanismos por trás de tais mudanças permanecem elusivas, principalmente no âmbito das interações celulares ocorrentes na medula óssea. Portanto, este trabalho buscou investigar como o envelhecimento afeta a regulação hematopoética no contexto de sua relação com as CTM medulares. Para esta pesquisa, foram utilizados camundongos machos isogênicos da linhagem C57BL/6, dividindoos em grupos conforme sua idade: jovens (3 ­ 5 meses) e idosos (18 ­ 19 meses). Foi realizada a caracterização do modelo através de aspectos físicos como consumo proteico, variação de peso, entre outros, seguido de avaliação bioquímica e hematológica. Adicionalmente, foram coletadas células medulares e, posteriormente, realizado o isolamento das CTMs. Para estudar a relação destas células com a hematopoese, foram realizados ensaios in vitro utilizando a linhagem celular leucêmica C1498 (TIB-49™, ATCC®) mantidas em contato com o sobrenadante das CTMs isoladas. Quanto aos parâmetros bioquímicos, os animais idosos apresentaram menores níveis de albumina, aspartato alanina transferase (ALT) e de triglicerídeos quando comparados aos animais jovens. Contrariamente, os animais idosos apresentaram um maior nível de colesterol. Na avaliação hematológica, foi constatado pelo hemograma que os animais idosos apresentaram valores comparáveis aos animais jovens, todavia, o mielograma mostrou menor celularidade geral, seguido de menor número de células da linhagem eritroide e maior número de precursores granulocíticos. Através da imunofenotipagem, foi revelado um maior número de CTHs e de precursores grânulosmonocíticos na medula de animais idosos quando comparado aos jovens, e uma menor frequência de progenitores linfoides. Na imunofenotipagem de sangue periférico de animais idosos houve uma redução no número de linfócitos B e de eritrócitos, e aumento na população de células natural killers. Na imunofenotipagem de CTMs, o marcador CD73 apresentou menor expressão nos animais idosos. Avaliando o secretoma destas células estromais, foram encontrados no sobrenadante de CTMs de animais idosos aumentos significativos nas concentrações de CXCL12 e SCF e redução de IL-11. No âmbito molecular, as CTMs de animais idosos apresentaram aumento na expressão de Akt1, Nos e Ppar-γ, e redução na expressão de Csf3 e Cdh2. Adicionalmente, quando comparado a ação das CTMs de animais idosos em relação as CTMs de animais jovens, observou-se que CTMs de animais idosos foram capazes de aumentar a expressão de Sox2, Pou5f1 e Nanog e diminuir a expressão de Cdkn1a de células da linhagem C1498. O sobrenadante de CTMs de animais idosos também resultou na maior proliferação e migração de células da linhagem C1498. Portanto, levando em consideração a importância das CTMs sobre a regulação do sistema hematopoético, pode-se concluir que, no envelhecimento, as CTMs criam um ambiente propício para a proliferação celular no qual a manutenção da pluripotência é estimulada, o que pode acarretar em uma desregulação do sítio hematopoético quando habitado por células malignas


Aging is a physiological process in which occurs a series of alterations in an organism that extend to a molecular level. It is a complex process that affects various tissues, one of them being the bone marrow, wherethrough the interactions of the hematopoietic stem cell (CTH) with its surrounding environment, including with the mesenchymal stem cell (CTM), hematopoiesis takes place. Although some aging-associated alterations in the bone marrow can be found described in the literature, the mechanisms behind said changes remain elusive, especially when regarding the cellular interactions present inside the bone marrow. Therefore, this research aimed to investigate how aging affects the regulation of hematopoiesis in the context of its interactions with bone marrow-derived CTMs. For this investigation, male isogenic C57BL/6 mice were used as animal models. These were separated in two groups according to their age: young (3 ­ 5 months) and aged (18 ­ 19 months). The animal models were characterized by their physical properties such as protein intake and weight variation, followed by biochemical and hematological evaluation. Bone marrow cells were obtained and identified through immunophenotyping, thus isolating different cell populations, including the CTMs. To study the relationship between these cells and hematopoiesis, in vitro assays were conducted utilizing the leukemic cell lineage C1498 (TIB-49™, ATCC®) maintained in contact with the supernatant of isolated CTMs. By their biochemical profile, aged mice showed lower levels of albumin, alanine-aspartate transferase (ALT) and triglycerides compared to the young group. In contrast, aged mice had a higher cholesterol level. Hematological evaluation by total blood count showed similar results between the two groups, however, the myelogram revealed that the aged animals had lower cellularity, with less frequent cells from the erythroid lineage, with an increase in granulocytic precursors. Through immunophenotyping, it was also revealed that aged mice have higher numbers of hematopoietic stem cells, while also being noted a reduced population of lymphoid progenitors. An increase in the granulomonocytic progenitors was also found. Immunophenotyping peripheral blood cells of aged mice revealed reduced numbers of B lymphocytes and erythrocytes, and an increased natural killer cell population. Additionally, the cell surface marker CD73 was found to be less expressed in aged mice CTMs. The secretome of these stromal cells obtained from aged mice showed higher levels of CXCL12 and SCF, and lower levels of IL-11when compared to the young counterparts. At a molecular level, CTMs obtained from aged mice expressed more Akt1, Nos and Ppar-γ, while the expression of Csf3 and Cdh2 was reduced. Additionally, when comparing the effects of aged mice CTMs with young mice CTMs, it was observed that the first expressed were capable of increasing the expression of Sox2, Pou5f1 and Nanog, while decreasing Cdkn1a expression in the C1498 cell lineage. The supernatant obtained from aged mice also favored the proliferation and cell migration of the C1498 cell line. Thus, considering the importance that CTMs have over the hematopoietic system, we can conclude that, in aging, CTMs create a special environment which favors cell proliferation and maintenance of pluripotency, which can result in a dysregulation of the hematopoietic tissue when malignant cells are present


Sujet(s)
Animaux , Mâle , Souris , Vieillissement/métabolisme , Cellules souches mésenchymateuses/classification , Hématopoïèse/génétique , Cellules souches hématopoïétiques/classification , Système hématopoïétique/malformations
2.
Nat Commun ; 9(1): 5180, 2018 12 05.
Article de Anglais | MEDLINE | ID: mdl-30518921

RÉSUMÉ

We previously demonstrated that an integrated XIST transgene can broadly repress one chromosome 21 in Down syndrome (DS) pluripotent cells. Here we address whether trisomy-silencing can normalize cell function and development sufficiently to correct cell pathogenesis, tested in an in vitro model of human fetal hematopoiesis, for which DS cellular phenotypes are best known. XIST induction in four transgenic clones reproducibly corrected over-production of megakaryocytes and erythrocytes, key to DS myeloproliferative disorder and leukemia. A contrasting increase in neural stem and iPS cells shows cell-type specificity, supporting this approach successfully rebalances the hematopoietic developmental program. Given this, we next used this system to extend knowledge of hematopoietic pathogenesis on multiple points. Results demonstrate trisomy 21 expression promotes over-production of CD43+ but not earlier CD34+/CD43-progenitors and indicates this is associated with increased IGF signaling. This study demonstrates proof-of-principle for this epigenetic-based strategy to investigate, and potentially mitigate, DS developmental pathologies.


Sujet(s)
Syndrome de Down/génétique , Syndrome de Down/thérapie , Extinction de l'expression des gènes , Thérapie génétique , Système hématopoïétique/malformations , ARN long non codant/génétique , Trisomie , Animaux , Chromosomes humains de la paire 21/génétique , Chromosomes humains de la paire 21/métabolisme , Syndrome de Down/métabolisme , Syndrome de Down/physiopathologie , Femelle , Hématopoïèse , Système hématopoïétique/métabolisme , Humains , Cellules souches pluripotentes induites/métabolisme , Mâle , Souris , ARN long non codant/métabolisme
3.
Mol Cell Biol ; 29(1): 229-40, 2009 Jan.
Article de Anglais | MEDLINE | ID: mdl-18936156

RÉSUMÉ

The Protection of telomeres 1 (POT1) protein is a single-stranded telomere binding protein that is essential for proper maintenance of telomere length. Disruption of POT1 function leads to chromosome instability and loss of cellular viability. Here, we show that targeted deletion of the mouse Pot1b gene results in increased apoptosis in highly proliferative tissues. In the setting of telomerase haploinsufficiency, loss of Pot1b results in depletion of germ cells and complete bone marrow failure due to increased apoptosis, culminating in premature death. Pot1b(-/-) mTR(+/-) hematopoietic progenitor and stem cells display markedly reduced survival potential in vitro. Accelerated telomere shortening, increased G overhang and elevated number of chromosome end-to-end fusions that initiate an ATR-dependent DNA damage response were also observed. These results indicate an essential role for Pot1b in the maintenance of genome integrity and the long-term viability of proliferative tissues in the setting of telomerase deficiency. Interestingly, these phenotypes closely resemble those found in the human disease dyskeratosis congenita (DC), an inherited syndrome characterized by bone marrow failure, hyperpigmentation, and nail dystrophy. We anticipate that this mouse will serve as a useful model to further understand the pathophysiology of DC.


Sujet(s)
Protéines du cycle cellulaire/métabolisme , Altération de l'ADN , Protéines de liaison à l'ADN/déficit , Dyskératose congénitale/génétique , Délétion de gène , Haploïdie , Protein-Serine-Threonine Kinases/métabolisme , Telomerase/déficit , Animaux , Protéines mutées dans l'ataxie-télangiectasie , Cellules de la moelle osseuse/enzymologie , Cellules de la moelle osseuse/anatomopathologie , Mort cellulaire , Prolifération cellulaire , Dyskératose congénitale/enzymologie , Système hématopoïétique/malformations , Système hématopoïétique/enzymologie , Système hématopoïétique/anatomopathologie , Souris , Souris knockout , Conformation d'acide nucléique , Spécificité d'organe , Phénotype , Analyse de survie , Telomerase/métabolisme , Télomère/composition chimique , Télomère/métabolisme
4.
Am J Pathol ; 173(6): 1693-701, 2008 Dec.
Article de Anglais | MEDLINE | ID: mdl-18988802

RÉSUMÉ

Mast cells participate in pathophysiological processes that range from antimicrobial defense to anaphylaxis and inflammatory arthritis. Much of the groundwork for the understanding of mast cells was established in mice that lacked mast cells through defects in either stem cell factor or its receptor, Kit. Among available strains, C57BL/6-Kit(W-sh) (W(sh)) mice are experimentally advantageous because of their background strain and fertility. However, the genetic inversion responsible for the W(sh) phenotype remains poorly defined, and its effects beyond the mast cell have been incompletely characterized. We report that W(sh) animals exhibit splenomegaly with expanded myeloid and megakaryocyte populations. Hematopoietic abnormalities extend to the bone marrow and are reflected by neutrophilia and thrombocytosis. In contrast, mast cell-deficient WBB6F1-Kit(W)/Kit(W-v) (W/W(v)) mice display mild neutropenia, but no changes in circulating platelet numbers. To help define the basis for the W(sh) phenotype, a "DNA walking" strategy was used to identify the precise location of the 3' breakpoint, which was found to reside 67.5 kb upstream of Kit. The 5' breakpoint disrupts corin, a cardiac protease responsible for the activation of atrial natriuretic peptide. Consistent with this result, transcription of full-length corin is ablated and W(sh) mice develop symptoms of cardiomegaly. Studies performed using mast cell-deficient strains must consider the capacity of associated abnormalities to either expose or compensate for the missing mast cell lineage.


Sujet(s)
Inversion chromosomique , Cardiopathies congénitales/génétique , Coeur/physiologie , Système hématopoïétique , Mastocytes/physiologie , Serine endopeptidases/métabolisme , Animaux , Système hématopoïétique/malformations , Système hématopoïétique/immunologie , Mâle , Mastocytes/cytologie , Souris , Souris de lignée C57BL , Phénotype , Protéines proto-oncogènes c-kit/génétique , Protéines proto-oncogènes c-kit/métabolisme , Serine endopeptidases/génétique , Rate/malformations , Rate/cytologie , Cellules souches/cytologie , Cellules souches/physiologie
5.
J Clin Invest ; 118(5): 1622-5, 2008 May.
Article de Anglais | MEDLINE | ID: mdl-18431512

RÉSUMÉ

HIV-associated hematological abnormalities involve all lineages of blood cells, thus implying that the virus impairs the function of early HSCs. However, the underlying mechanisms of this defect are unknown, particularly since HSCs are largely resistant to HIV-1 infection. In this issue of the JCI, Prost and colleagues show that the viral accessory protein Negative factor (Nef) plays a potentially critical role in the pathogenesis of HIV/SIV-associated hematopoietic dysfunction by affecting the clonogenic potential of HSCs (see the related article beginning on page 1765). Soluble Nef induces PPARgamma in uninfected HSCs, thereby suppressing the expression of STAT5A and STAT5B, two factors necessary for proper HSC function. The identification of this novel activity of extracellular Nef defines a new mechanism of HIV/SIV pathogenesis and suggests that approaches aimed at increasing STAT5A and STAT5B expression may be considered in HIV-infected individuals with prominent hematological abnormalities. The results also raise the question of whether dysregulation of hematopoiesis by extracellular Nef plays a role in the development of T cell immunodeficiency and the high levels of chronic immune activation associated with AIDS.


Sujet(s)
Infections à VIH/physiopathologie , Système hématopoïétique , Produits du gène nef du virus de l'immunodéficience humaine/métabolisme , Animaux , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/physiologie , Système hématopoïétique/malformations , Système hématopoïétique/physiologie , Humains , Récepteur PPAR gamma/métabolisme , Facteur de transcription STAT-5/génétique , Facteur de transcription STAT-5/métabolisme , Syndrome d'immunodéficience acquise du singe/physiopathologie , Virus de l'immunodéficience simienne/métabolisme , Produits du gène nef du virus de l'immunodéficience humaine/génétique
6.
Exp Hematol ; 35(9): 1421-8, 2007 Sep.
Article de Anglais | MEDLINE | ID: mdl-17761289

RÉSUMÉ

OBJECTIVE: Members of the hox family of homeodomain-containing transcription factors, including hoxa9, hoxb3, and hoxb4 play an important role in the regulation of differentiation, proliferation and self-renewal of hematopoietic stem and progenitor cells. Lack-of-function studies using hoxa9, hoxb4, or hoxb3/hoxb4 null mice demonstrate that all these mutations compromise the repopulating ability of hematopoietic stem cells (HSC), implying similar functions of each of these genes in hematopoiesis. Because cross regulation and cooperativity are known features of hox proteins, we investigated mice with a compound deficiency in hoxa9, hoxb3 and hoxb4 (hoxa9/b3/b4) for evidence of synergy between these genes in hematopoiesis. MATERIALS AND METHODS: Hoxa9/b3/b4 were generated by mating the hoxb3/hoxb4 null mice with the hoxa9 null strain and HSC function was measured by competitive repopulating assay and by immunophenotype using fluorescence-activated cell sorting. RESULTS: Our findings demonstrate that the hoxa9/b3/b4 null mice are smaller in body weight, and display a marked reduction in spleen size and cellularity compared to control mice. The numbers of colony-forming unit (CFU)-granulocyte macrophage and CFU-spleen progenitor colonies were normal but hoxa9/b3/b4 null bone marrow contained increased numbers of immunophenotypic HSC (Lin(-), c-kit(+), Sca-1(+), CD150(+)). However the reconstitution defect in hoxa9 null HSC was not enhanced further in the hoxa9/b3/b4 null HSC. CONCLUSION: These findings demonstrate overlapping functions of hoxa9, hoxb3, and hoxb4 in hematopoietic cells, and emphasize an important role for these transcription factors for regulation of HSC proliferation. However, none of these hox proteins is absolutely essential for generation or maintenance of all major blood lineages.


Sujet(s)
Système hématopoïétique/malformations , Protéines à homéodomaine/physiologie , Facteurs de transcription/physiologie , Animaux , Poids , Lignage cellulaire , Système hématopoïétique/anatomopathologie , Protéines à homéodomaine/génétique , Agranulocytes/anatomopathologie , Souris , Souris knockout , Mutation , Taille d'organe , Rate/anatomie et histologie , Rate/cytologie , Facteurs de transcription/génétique
7.
Mol Cell Biol ; 24(4): 1570-81, 2004 Feb.
Article de Anglais | MEDLINE | ID: mdl-14749373

RÉSUMÉ

The BH3-only members of the Bcl-2 protein family are essential for initiation of programmed cell death and stress-induced apoptosis. We have determined the expression pattern in mice of the BH3-only protein Bik, also called Blk or Nbk, and examined its physiological function by gene targeting. We found that Bik is expressed widely in the hematopoietic compartment and in endothelial cells of the venous but not arterial lineages. Nevertheless, its loss did not increase the numbers of such cells in mice or protect hematopoietic cells in vitro from apoptosis induced by cytokine withdrawal or diverse other cytotoxic stimuli. Moreover, whereas loss of the BH3-only protein Bim rescued mice lacking the prosurvival protein Bcl-2 from fatal polycystic kidney disease and lymphopenia, loss of Bik did not. These results indicate that any function of Bik in programmed cell death and stress-induced apoptosis must overlap that of other BH3-only proteins.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Apoptose , Protéines de transport/métabolisme , Cellules endothéliales/métabolisme , Système hématopoïétique/métabolisme , Protéines mitochondriales , Protéines proto-oncogènes c-bcl-2/métabolisme , Animaux , Protéines régulatrices de l'apoptose , Protéines de transport/génétique , Cellules endothéliales/cytologie , Femelle , Délétion de gène , Analyse de profil d'expression de gènes , Gènes rapporteurs/génétique , Hématopoïèse , Système hématopoïétique/malformations , Système hématopoïétique/cytologie , Rein/malformations , Souris , Souris knockout , Néovascularisation physiologique , Protéines proto-oncogènes c-bcl-2/déficit , Protéines proto-oncogènes c-bcl-2/génétique , ARN messager/génétique , ARN messager/métabolisme
8.
Development ; 128(24): 5173-80, 2001 Dec.
Article de Anglais | MEDLINE | ID: mdl-11748152

RÉSUMÉ

Phospholipase C-gamma1 (PLC-gamma1) is involved in a variety of intracellular signaling via many growth factor receptors and T-cell receptor. To explore the role of PLC-gamma1 in vivo, we generated the PLC-gamma1-deficient (plc-gamma1(-/-)) mice, which died of growth retardation at embryonic day 8.5-9.5 in utero. Therefore, we examined plc-gamma1(-/-) chimeric mice generated with plc-gamma1(-/-) embryonic stem (ES) cells for further study. Pathologically, plc-gamma1(-/-) chimeras showed multicystic kidney due to severe renal dysplasia and renal tube dilation. Flow cytometric analysis and glucose phosphate isomerase assay revealed very few hematopoietic cells derived from the plc-gamma1(-/-) ES cells in the mutant chimeras. However, differentiation of plc-gamma1(-/-) ES cells into erythrocytes and monocytes/macrophages in vitro was observed to a lesser extent compared with control wild-type ES cells. These data suggest that PLC-gamma1 plays an essential role in the renal development and hematopoiesis in vivo.


Sujet(s)
Hématopoïèse/génétique , Isoenzymes/déficit , Dysplasie rénale multikystique/étiologie , Type C Phospholipases/déficit , Animaux , Antigènes CD/isolement et purification , Différenciation cellulaire , Chimère , Embryon de mammifère/cytologie , Système hématopoïétique/malformations , Isoenzymes/génétique , Tissu lymphoïde/malformations , Souris , Souches mutantes de souris , Dysplasie rénale multikystique/génétique , Phospholipase C gamma , Famille des molécules de signalisation de l'activation des lymphocytes , Cellules souches/cytologie , Type C Phospholipases/génétique
9.
Eur J Immunogenet ; 27(1): 29-33, 2000 Feb.
Article de Anglais | MEDLINE | ID: mdl-10651848

RÉSUMÉ

The McLeod syndrome is a rare X-linked recessive disorder characterized by blood group, neuromuscular and haematopoietic abnormalities. It is caused by XK gene defects and may include large deletions in the Xp21 region. Analysis of three unrelated McLeod patients for the presence of the XK, DMD, CYBB, ETX1, RPGR and OTC loci, as well as for the DXS709 marker, revealed deletions from the 39th exon of DMD to the ETX1 locus (patient Be), from the XK to RPGR loci (patient Bi) and from the XK to CYBB loci (patient Lh). All three patients normally expressed the Lutheran (Lu) red cell antigens, thus excluding the interval between the RPGR and DMD genes as site of the XS locus, previously mapped to the Xp21.2-Xq21.1 region and thought to regulate the expression of the LU blood group gene on chromosome 19.


Sujet(s)
Délétion de gène , Système hématopoïétique/malformations , Maladies neuromusculaires/génétique , Chromosome X , Allèles , Antigènes de groupe sanguin/génétique , Technique de Southern , Exons , Cytométrie en flux , Gènes récessifs , Humains , Réaction de polymérisation en chaîne , Syndrome
10.
Article de Anglais | MEDLINE | ID: mdl-10895256

RÉSUMÉ

The antigens of the Kell blood group system are carried on a 93 kDa type II glycoprotein encoded by a single gene on chromosome 7 at 7q33. XK is a 50.9 kDa protein that traverses the membrane ten times and derives from a single gene on the X chromosome at Xp21. A single disulphide bond, Kell Cys 72-XK Cys 347, links Kell to XK. The Kell component of the Kell/XK complex is important in transfusion medicine since it is a highly polymorphic protein, carrying over 23 different antigens, that can cause severe reactions if mismatched blood is transfused and in pregnant mothers antibodies to Kell may elicit serious fetal and neonatal anaemia. The different Kell phenotypes are all caused by base mutations leading to single amino acid substitutions. By contrast the XK component carries a single blood group antigen, termed Kx. The physiological functions of Kell and XK have not been fully elucidated but Kell is a zinc endopeptidase with endothelin-3-converting enzyme activity and XK has the structural characteristics of a membrane transporter. Lack of Kx, the McLeod phenotype, is associated with red cell acanthocytosis, elevated levels of serum creatine phosphokinase and late onset forms of muscular and neurological defects.


Sujet(s)
Systèmes de transport d'acides aminés neutres , Protéines de transport/génétique , Système hématopoïétique/malformations , Système Kell , Protéines membranaires/génétique , Maladies neuromusculaires/génétique , Acanthocytes/anatomopathologie , Protéines de transport/sang , Liaison génétique , Génotype , Humains , Système Kell/sang , Système Kell/composition chimique , Système Kell/génétique , Protéines membranaires/sang , Maladies neuromusculaires/sang , Phénotype , Syndrome , Chromosome X
11.
Genes Dev ; 8(7): 757-69, 1994 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-7926765

RÉSUMÉ

The bmi-1 proto-oncogene has been implicated in B-cell lymphomagenesis in E mu-myc transgenic mice. Distinct domains of the Bmi-1 protein are highly conserved within the Drosophila protein Posterior Sex Combs, a member of the Polycomb group involved in maintaining stable repression of homeotic genes during development. We have inactivated the bmi-1 gene in the germ line of mice by homologous recombination in ES cells. Null mutant mice display three phenotypic alterations: (1) a progressive decrease in the number of hematopoietic cells and an impaired proliferative response of these cells to mitogens; (2) neurological abnormalities manifested by an ataxic gait and sporadic seizures; and (3) posterior transformation, in most cases along the complete anteroposterior axis of the skeleton. The observations indicate that Mbi-1 plays an important role in morphogenesis during embryonic development and in hematopoiesis throughout pre- and postnatal life. Furthermore, these data provide the first evidence of functional conservation of a mammalian Polycomb group homolog.


Sujet(s)
Encéphale/malformations , Système hématopoïétique/malformations , Protéines nucléaires/physiologie , Protéines proto-oncogènes/physiologie , Protéines de répression , Rachis/malformations , Animaux , Ataxie/génétique , Cellules de la moelle osseuse , Développement embryonnaire et foetal , Femelle , Régulation de l'expression des gènes au cours du développement , Ciblage de gène , Mutation germinale/physiologie , Système hématopoïétique/cytologie , Activation des lymphocytes , Mâle , Souris , Souris transgéniques , Mitogènes/pharmacologie , Protéines nucléaires/génétique , Complexe répresseur Polycomb-1 , Protéines proto-oncogènes/génétique , Proto-oncogènes , Recombinaison génétique , Côtes/malformations , Crises épileptiques/génétique , Rate/cytologie , Thymus (glande)/cytologie
12.
Acta méd. colomb ; 9(2): 76-81, 1984. ilus, tab
Article de Espagnol | LILACS | ID: lil-292737

RÉSUMÉ

Se presentan dos nuevos casos de disqueratosis congénita, las primeras en la literatura médica, así como el cuarto y quinto casos de sexo femenino, haciendo hincapié en sus características clínicas, asociación y relación con neoplasia maligna y alteraciones hematopoyéticas pancitopénicas, que la convierten en una enfermedad dermatológica potencialmente letal


Sujet(s)
Humains , Femelle , Adulte , Kératose/génétique , Système hématopoïétique/malformations , Système hématopoïétique/traumatismes , Système hématopoïétique/anatomopathologie , Maladies de la peau/diagnostic , Maladies de la peau/étiologie , Maladies de la peau/génétique
15.
Proc Natl Acad Sci U S A ; 68(1): 124-6, 1971 Jan.
Article de Anglais | MEDLINE | ID: mdl-5276282

RÉSUMÉ

Young New Zealand Black (NZB) mice manifested extremely high resistance to the lethal effects of acute exposures to ionizing radiation, with a dose necessary to kill 50% of the animals within 30 days, LD(50(30)), of 964 roentgens (R) at 30 days of age and of 856 R for 90-day-old mice. In contrast, Coombs' positive 9-month-old NZB mice (with low primary immune response) were highly susceptible (LD(50(30)) = 543 R), possibly because of anemia-stimulated erythropoiesis leading to a depletion of stem cells. The radiation resistance of young NZB mice, combined with previous observations of their immunologic hyper-responsiveness, support the concept that NZB mice possess an unusually large pool of hematopoietic stem cells, an abnormality which may predispose them to the development of autoimmune disease and neoplasia.


Sujet(s)
Maladies auto-immunes/étiologie , Souris , Effets des rayonnements , Maladies des rongeurs/étiologie , Animaux , Moelle osseuse/effets des radiations , Cellules de la moelle osseuse , Test de Coombs , Système hématopoïétique/malformations , Tumeurs radio-induites/immunologie , Lésions radiques expérimentales/complications , Lésions radiques expérimentales/mortalité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...