Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Comput Biol Chem ; 113: 108209, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39260229

RESUMO

Fusion proteins have the potential to become the new norm for targeted therapeutic treatments. Highly specific payload delivery can be achieved by combining custom targeting moieties, such as VHH domains, with active parts of proteins that have a particular activity not naturally targeted to the intended cells. Conversely, novel drug products may make use of the highly specific targeting properties of naturally occurring proteins and combine them with custom payloads. When designing such a product, there is rarely a known structure for the final construct which makes it difficult to assess molecular behaviour that may ultimately impact therapeutic outcome. Considering the time and cost of expressing a construct, optimising the purification procedure, obtaining sufficient quantities for biophysical characterisation, and performing structural studies in vitro, there is an enormous benefit to conduct in silico studies ahead of wet lab work. By following a repeatable, streamlined, and fast workflow of molecular dynamics assessment, it is possible to eliminate low-performing candidates from costly experimental work. There are, however, many aspects to consider when designing a novel fusion protein and it is crucial not to overlook some elements. In this work, we suggest a set of user-friendly, open-source methods which can be used to screen fusion protein candidates from the sequence alone. We used the light chain and translocation domain of botulinum toxin A (BoNT/A) fused with a selected VHH domain, termed here LC-HN-VHH, as a case study for a general approach to designing, modelling, and simulating fusion proteins. Its behaviour in silico correlated well with initial in vitro work, with SEC HPLC showing multiple protein states in solution and a dynamic protein shifting between these states over time without loss of material.

2.
Leukemia ; 31(6): 1415-1422, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27840422

RESUMO

Activating mutations, such as E76K and D61Y, in PTPN11 (SHP2), a protein tyrosine phosphatase implicated in multiple cell signaling processes, are associated with 35% of patients with juvenile myelomonocytic leukemia (JMML), an aggressive childhood myeloproliferative neoplasm (MPN). Here we show that the interaction between leukemia-associated mutant Shp2 and Gab2, a scaffolding protein important for cytokine-induced PI3K/Akt signaling, was enhanced, and that the mTOR pathway was elevated in Ptpn11E76K/+ leukemic cells. Importantly, MPN induced by the Ptpn11E76K/+ mutation was markedly attenuated in Ptpn11E76K/+/Gab2-/- double mutant mice-overproduction of myeloid cells was alleviated, splenomegaly was diminished and myeloid cell infiltration in nonhematopoietic organs was decreased in these double mutants. Excessive myeloid differentiation of stem cells was also normalized by depletion of Gab2. Acute leukemia progression of MPN was reduced in the double mutant mice and, as such, their survival was much prolonged. Furthermore, treatment of Ptpn11E76K/+ mice with Rapamycin, a specific and potent mTOR inhibitor, mitigated MPN phenotypes. Collectively, this study reveals an important role of the Gab2/PI3K/mTOR pathway in mediating the pathogenic signaling of the PTPN11 gain-of-function mutations and a therapeutic potential of Rapamycin for PTPN11 mutation-associated JMML.


Assuntos
Mutação , Transtornos Mieloproliferativos/etiologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosfoproteínas/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antibióticos Antineoplásicos/farmacologia , Camundongos , Camundongos Knockout , Transtornos Mieloproliferativos/tratamento farmacológico , Transtornos Mieloproliferativos/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfoproteínas/genética , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/genética
3.
Mol Psychiatry ; 16(6): 672-84, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21403673

RESUMO

Calcyon regulates activity-dependent internalization of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) glutamate receptors and long-term depression of excitatory synapses. Elevated levels of calcyon are consistently observed in brains from schizophrenic patients, and the calcyon gene is associated with attention-deficit hyperactivity disorder. Executive function deficits are common to both disorders, and at least for schizophrenia, the etiology appears to involve both heritable and neurodevelopmental factors. Here, we show with calcyon-overexpressing Cal(OE) transgenic mice that lifelong calcyon upregulation impairs executive functions including response inhibition and working memory, without producing learning and memory deficits in general. As response inhibition and working memory, as well as the underlying neural circuitry, continue to mature into early adulthood, we functionally silenced the transgene during postnatal days 28-49, a period corresponding to adolescence. Remarkably, the response inhibition and working memory deficits including perseverative behavior were absent in adult Cal(OE) mice with the transgene silenced in adolescence. Suppressing the calcyon transgene in adulthood only partially rescued the deficits, suggesting calcyon upregulation in adolescence irreversibly alters development of neural circuits supporting mature response inhibition and working memory. Brain regional immunoblots revealed a prominent downregulation of AMPA GluR1 subunits in hippocampus and GluR2/3 subunits in hippocampus and prefrontal cortex of the Cal(OE) mice. Silencing the transgene in adolescence prevented the decrease in hippocampal GluR1, further implicating altered fronto-hippocampal connectivity in the executive function deficits observed in the Cal(OE) mice. Treatments that mitigate the effects of high levels of calcyon during adolescence could preempt adult deficits in executive functions in individuals at risk for serious mental illness.


Assuntos
Inibição Psicológica , Proteínas de Membrana/metabolismo , Transtornos da Memória/fisiopatologia , Memória de Curto Prazo/fisiologia , Regulação para Cima/fisiologia , Animais , Animais Recém-Nascidos , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/genética , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Modelos Animais de Doenças , Doxiciclina/metabolismo , Doxiciclina/farmacologia , Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/fisiologia , Medo/efeitos dos fármacos , Medo/fisiologia , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Proteínas de Membrana/genética , Transtornos da Memória/genética , Camundongos , Camundongos Transgênicos , Receptores de AMPA/metabolismo , Percepção Espacial/efeitos dos fármacos , Percepção Espacial/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
4.
Leukemia ; 24(8): 1397-405, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20535152

RESUMO

Signal transducer and activator of transcription-5 (STAT5) is a critical transcription factor for normal hematopoiesis and its sustained activation is associated with hematologic malignancy. A persistently active mutant of STAT5 (STAT5a(S711F)) associates with Grb2-associated binding protein 2 (Gab2) in myeloid leukemias and promotes growth in vitro through AKT activation. Here we have retrovirally transduced wild-type or Gab2(-/-) mouse bone marrow cells expressing STAT5a(S711F) and transplanted into irradiated recipient mice to test an in vivo myeloproliferative disease model. To target Gab2-independent AKT/mTOR activation, we treated wild-type mice separately with rapamycin. In either case, mice lacking Gab2 or treated with rapamycin showed attenuated myeloid hyperplasia and modestly improved survival, but the effects were not cytotoxic and were reversible. To improve on this approach, we combined in vitro targeting of STAT5-mediated AKT/mTOR using rapamycin with inhibition of the STAT5 direct target genes bcl-2 and bcl-X(L) using ABT-737. Striking synergy with both drugs was observed in mouse BaF3 cells expressing STAT5a(S711F), TEL-JAK2 or BCR-ABL and in the relatively single agent-resistant human BCR-ABL-positive K562 cell line. Therefore, targeting distinct STAT5-mediated survival signals, for example, bcl-2/bcl-X(L) and AKT/mTOR may be an effective therapeutic approach for human myeloproliferative neoplasms.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Compostos de Bifenilo/farmacologia , Transtornos Mieloproliferativos/patologia , Nitrofenóis/farmacologia , Fator de Transcrição STAT5/fisiologia , Sirolimo/farmacologia , Sulfonamidas/farmacologia , Animais , Antibióticos Antineoplásicos/administração & dosagem , Compostos de Bifenilo/administração & dosagem , Linhagem Celular Tumoral , Sobrevivência Celular , Citometria de Fluxo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Transtornos Mieloproliferativos/fisiopatologia , Nitrofenóis/administração & dosagem , Piperazinas/administração & dosagem , Piperazinas/farmacologia , Fator de Transcrição STAT5/efeitos dos fármacos , Sirolimo/administração & dosagem , Sulfonamidas/administração & dosagem
5.
Bone Marrow Transplant ; 41(5): 465-72, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18246116

RESUMO

Allogeneic hematopoietic stem cell (HSC) transplantation is a life-saving procedure for hematopoietic malignancies, marrow failure syndromes and hereditary immunodeficiency disorders. However, wide application of this procedure is limited by availability of suitable human leucocyte antigen (HLA)-matched adult donors. Umbilical cord blood (UCB) has been increasingly used as an alternative HSC source for patients lacking matched-HSC donors. The clinical experience of using UCB transplantation to treat pediatric acute leukemias has already shown that higher-level HLA-mismatched UCB can be equally as good as or even better than matched HSC. Recently, large registries and multiple single institutional studies conclusively demonstrated that UCB is an acceptable source of HSCs for adult acute leukemia patients who lack HLA-matched donors. These studies will impact the future clinical allogeneic stem cell transplantation for acute myeloid leukemia (AML), which is the most common acute leukemia in adults. UCB has unique advantages of easy procurement, absence of risk to donors, low risk of transmitting infections, immediate availability, greater tolerance of HLA disparity and lower-than-expected incidence of severe graft-versus-host disease. These features of UCB permit successful transplantation available to almost every patient who needs it. We anticipate that using UCB as a HSC source for allogeneic transplantation for adult AML will increase dramatically over the next 5 years, by expanding the available allogeneic donor pool. Clinical studies are needed with focus on disease-specific UCB transplantation outcomes, including AML, acute lymphoblastic leukemia, and lymphoma.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Leucemia Mieloide Aguda/terapia , Condicionamento Pré-Transplante/métodos , Adulto , Bancos de Sangue , Sangue Fetal/fisiologia , Teste de Histocompatibilidade , Humanos
6.
Nat Med ; 7(9): 1028-34, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11533706

RESUMO

Stem cells from bone marrow, skeletal muscle and possibly other tissues can be identified by the 'side-population' (SP) phenotype. Although it has been assumed that expression of ABC transporters is responsible for this phenotype, the specific molecules involved have not been defined. Here we show that expression of the Bcrp1 (also known as Abcg2 murine/ABCG2 human) gene is a conserved feature of stem cells from a wide variety of sources. Bcrp1 mRNA was expressed at high levels in primitive murine hematopoietic stem cells, and was sharply downregulated with differentiation. Enforced expression of the ABCG2 cDNA directly conferred the SP phenotype to bone-marrow cells and caused a reduction in maturing progeny both in vitro and in transplantation-based assays. These results show that expression of the Bcrp1/ABCG2 gene is an important determinant of the SP phenotype, and that it might serve as a marker for stem cells from various sources.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas Inflamatórias de Macrófagos , Glicoproteínas de Membrana , Proteínas Associadas à Resistência a Múltiplos Medicamentos , Proteínas de Neoplasias , Células-Tronco/fisiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Biomarcadores , Células da Medula Óssea/fisiologia , Células Cultivadas , Quimiocinas CC , Citocinas/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Mutantes , Proteínas Ribossômicas/metabolismo , Células-Tronco/citologia , Tetraspanina 29 , Transplantes
7.
Chem Biol Interact ; 130-132(1-3): 261-73, 2001 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-11306050

RESUMO

Toxic medium chain length alkanals, alkenals, and 4-hydroxyalkenals that are generated during lipid peroxidation are potential substrates for aldehyde dehydrogenase (ALDH) isoforms. We have developed transgenic cell lines to examine the potential for either human ALDH1A1 or ALDH3A1 to protect against damage mediated by these toxic aldehydes. Using crude cytosols from stably transfected cell lines, these aldehydes were confirmed to be excellent substrates for ALDH3A1, but were poorly oxidized by ALDH1A1. Expression of ALDH3A1 by stable transfection in V79 cells conferred a high level of protection against growth inhibition by the medium-chain length aldehyde substrates with highest substrate activity, including hexanal, trans-2-hexenal, trans-2-octenal, trans-2-nonenal, and 4-hydroxy-2-nonenal (HNE). This was reflected in a parallel ability of ALDH3A1 to prevent depletion of glutathione by these aldehydes. Expression of hALDH3 completely blocked the potent induction of apoptosis by HNE in both V79 cells and in a RAW 264.7 murine macrophage cell line, consistent with the observed total prevention of HNE-protein adduct formation. Structure-activity studies indicated that the rank order of potency for the contributions of HNE functional groups to toxicity was aldehyde >/=C2=C3 double bond>>C4-hydroxyl group. Oxidation of the aldehyde moiety of HNE to a carboxyl by ALDH3A1 expressed in stably transfected cell lines drastically reduced its potency for growth inhibition and apoptosis induction. In contrast, ALDH1A1 expression provided only moderate protection against trans-2-nonenal (t2NE), and none against the other six-nine carbon aldehydes. Neither ALDH1A1 nor ALDH3A1 conferred any protection against acrolein, acetaldehyde, or chloroacetaldehyde. A small degree of protection against malondialdehyde was afforded by ALDH1A1, but not ALDH3A1. Paradoxically, cells expressing ALDH3A1 were 1.5-fold more sensitive to benzaldehyde toxicity than control V79 cells. These studies demonstrate that expression of class 3 ALDH, but not class 1 ALDH, can be an important determinant of cellular resistance to toxicity mediated by aldehydes of intermediate chain length that are produced during lipid peroxidation.


Assuntos
Acetaldeído/análogos & derivados , Aldeído Desidrogenase/genética , Aldeído Desidrogenase/metabolismo , Aldeídos/farmacocinética , Aldeídos/toxicidade , Isoenzimas/genética , Isoenzimas/metabolismo , Acetaldeído/toxicidade , Acroleína/toxicidade , Família Aldeído Desidrogenase 1 , Alquilação , Animais , Apoptose/efeitos dos fármacos , Benzaldeídos/toxicidade , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Cricetinae , Resistência a Medicamentos , Humanos , Inativação Metabólica , Peroxidação de Lipídeos , Proteínas/metabolismo , Ratos , Retinal Desidrogenase , Transfecção
8.
Mol Cell Biol ; 21(2): 678-89, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11134353

RESUMO

Jak3-deficient mice display vastly reduced numbers of lymphoid cells. Thymocytes and peripheral T cells from Jak3-deficient mice have a high apoptotic index, suggesting that Jak3 provides survival signals. Here we report that Jak3 regulates T lymphopoiesis at least in part through its selective regulation of Bax and Bcl-2. Jak3-deficient thymocytes express elevated levels of Bax and reduced levels of Bcl-2 relative to those in wild-type littermates. Notably, up-regulation of Bax in Jak3-deficient T cells is physiologically relevant, as Jak3 Bax double-null mice have marked increases in thymocyte and peripheral T-cell numbers. Rescue of T lymphopoiesis by Bax loss was selective, as mice deficient in Jak3 plus p53 or in Jak3 plus Fas remained lymphopenic. However, Bax loss failed to restore proper ratios of peripheral CD4/CD8 T cells, which are abnormally high in Jak3-null mice. Transplantation into Jak3-deficient mice of Jak3-null bone marrow transduced with a Bcl-2-expressing retrovirus also improved peripheral T-cell numbers and restored the ratio of peripheral CD4/CD8 T cells to wild-type levels. The data support the concepts that Jak kinases regulate cell survival through their selective and cell context-dependent regulation of pro- and antiapoptotic Bcl-2 family proteins and that Bax and Bcl-2 play distinct roles in T-cell development.


Assuntos
Regulação da Expressão Gênica , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas/genética , Linfócitos T/citologia , Animais , Apoptose , Relação CD4-CD8 , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular , Sobrevivência Celular , Citometria de Fluxo , Imunofluorescência , Deleção de Genes , Marcação In Situ das Extremidades Cortadas , Janus Quinase 3 , Camundongos , Camundongos Knockout , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Linfócitos T/metabolismo , Timo/citologia , Timo/crescimento & desenvolvimento , Proteína X Associada a bcl-2
9.
Hum Gene Ther ; 11(17): 2353-64, 2000 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-11096440

RESUMO

Janus kinase 3 (JAK3) is an essential component of cytokine receptor signal transduction pathways required for normal lymphocyte development and function. JAK3 deficiency in both mice and humans results in severe combined immunodeficiency (SCID) and increased susceptibility to opportunistic infections. We have previously shown that JAK3 gene transfer into irradiated recipients could restore immune function. However, since this toxic conditioning would be undesirable for infants in a clinical application, we have tested whether immune function could be restored in nonmyeloablated JAK3-deficient (-/-) mice. Murine JAK3 retroviral vectors were transduced into hematopoietic stem cells from the livers of newborn JAK3(-/-) mice. These cells were then injected intraperitoneally into nonirradiated JAK3(-/-) neonates. Transduced cells were detectable in these mice at time points 4 to 6 months after injection and resulted in significant correction of T and B lymphocyte numbers and circulating immunoglobulin (Ig) levels. After immune challenge with a dose of influenza A virus that was lethal to nonmanipulated JAK3(-/-) mice, mice injected with transduced cells showed development of circulating virus-specific IgG and enhanced survival. This work shows that the large selective advantage for JAK3-corrected lymphoid cells may be sufficient to overcome the need for myeloablative conditioning in JAK3 gene therapy protocols.


Assuntos
Terapia Genética/métodos , Células-Tronco Hematopoéticas/fisiologia , Linfócitos/fisiologia , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Imunodeficiência Combinada Severa/terapia , Animais , Animais Recém-Nascidos , Formação de Anticorpos , Transplante de Células , Vetores Genéticos , Vírus da Influenza A/patogenicidade , Janus Quinase 3 , Fígado/citologia , Camundongos , Camundongos Mutantes , Células Mieloides/fisiologia , Infecções por Orthomyxoviridae/imunologia , Fenótipo , Retroviridae/genética , Seleção Genética , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/patologia , Taxa de Sobrevida
10.
Blood ; 96(6): 2277-83, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10979977

RESUMO

The cytokines interleukin 7 (IL-7) and interleukin 4 (IL-4) regulate lymphoid differentiation and function and activate the transcription factor Stat5. Using mice deficient for the 2 highly related transcription factors, Stat5a and Stat5b (Stat5a/b(-/-)), we investigated the role of Stat5 for B-cell differentiation, expansion, and function. Peripheral blood B cells of Stat5-deficient mice are significantly reduced, but no proliferation defects in response to various mitogenic stimuli are found. Also, IgM and IgG1 antibody production and immunoglobulin class switching are not affected. Pre- and pro-B cells of Stat5-deficient animals were found to have reduced responses to IL-7. Pro- and pre-B cells are the target cells of the abl oncogene and numerous studies have suggested that Stat5a/b is essential for transformation by derivatives of the Abelson (abl) gene. To assess the role of Stat5a/b in transformation, we have evaluated the ability of various abl derivatives to transform cells from Stat5a/b-deficient mice in vitro or in vivo. We demonstrate that the absence of Stat5a/b is not essential for the induction of lymphoid or myeloid tumors in vivo or on the ability to transform bone marrow cells in vitro.


Assuntos
Linfócitos B/patologia , Linfócitos B/fisiologia , Transformação Celular Neoplásica/genética , Proteínas de Ligação a DNA/genética , Proteínas de Fusão bcr-abl/genética , Interleucina-7/genética , Proteínas do Leite , Transativadores/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Genes abl , Interleucina-7/farmacologia , Camundongos , Fator de Transcrição STAT5
11.
Blood ; 96(3): 902-9, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10910903

RESUMO

The human multidrug resistance-1 (MDR1) gene product, P-glycoprotein (P-gp), is well known for its ability to confer drug resistance; however, recent evidence suggests that P-gp expression can have more general effects on cellular development. In support of this idea, it was previously shown that retroviral-mediated MDR1 expression in murine bone marrow cells resulted in the expansion of stem cells in culture and in the development of a myeloproliferative syndrome in transplanted mice. It is now reported that MDR1-mediated stem cell expansion is associated with an increase in side population (SP) stem cells, defined by Hoechst dye staining. Transduction of murine bone marrow cells with an MDR1 retroviral vector resulted in an almost 2 log increase in SP cell numbers over 12 days in culture, whereas there was a rapid loss of SP cells from control cultures. Stem cell amplification was not limited to ex vivo expansion cultures but was also evident when MDR1-transduced cells were directly transplanted into irradiated mice. In these cases, stem cell expansion was associated with relatively high vector copy numbers in stem cell clones. As previously reported, some cases were associated with a characteristic myeloproliferative syndrome. A functionally inactive MDR1 mutant cDNA was used to show that P-gp pump function was required both for amplification of phenotypically defined SP cells and functionally defined repopulating cells. These studies further support the concept that ABC transporter function can have important effects on hematopoietic stem cell development.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Técnicas de Transferência de Genes , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Contagem de Células Sanguíneas , Diferenciação Celular/genética , Divisão Celular/genética , Expressão Gênica , Vetores Genéticos , Humanos , Camundongos , Retroviridae
12.
Ann N Y Acad Sci ; 872: 125-40; discussion 140-1, 1999 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-10372117

RESUMO

Ex vivo expansion of hematopoietic stem cells would be useful for bone marrow transplantation and gene therapy applications. Toward this goal, we have investigated whether retrovirally-transduced murine stem cells could be expanded in culture with hematopoietic cytokines. Bone marrow cells were transduced with retroviral vectors expressing either the human multidrug resistance 1 gene (HaMDR1), a variant of human dihydrofolate reductase (HaDHFR), or both MDR1 and DHFR in an internal ribosomal entry site (IRES)-containing bicistronic vector (HaMID). Cells were then expanded for 15 days in cultures stimulated with interleukin (IL)-3, IL-6, and stem cell factor. When very low marrow volumes were injected into lethally irradiated recipient mice, long-term reconstitution with 100% donor cells was seen in all mice injected with HaMDR1- or HaMID-transduced cells. By contrast, engraftment with HaDHFR- or mock-transduced cells ranged from partial to undetectable despite injection of significantly larger marrow volumes. In addition, mice transplanted with expanded HaMDR1- or HaMID-transduced stem cells developed a myeloproliferative disorder that was characterized by an increase in abnormal peripheral blood leukocytes. These results show that MDR1-transduced stem cells can be expanded in vitro with hematopoietic cytokines, but indicate that an increased stem cell division frequency can lead to stem cell damage.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Retroviridae , Transfecção/métodos , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/sangue , Animais , Animais Recém-Nascidos , Diferenciação Celular , Divisão Celular , Células Cultivadas , Humanos , Leucócitos/citologia , Leucócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Modelos Biológicos
13.
Immunity ; 10(2): 249-59, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10072077

RESUMO

Many cytokines activate two highly homologous Stat proteins, 5a and 5b. Mice deficient in both genes lack all growth hormone and prolactin functions but retain functions associated with cytokines such as erythropoietin. Here, we demonstrate that, while lymphoid development is normal, Stat5a/b mutant peripheral T cells are profoundly deficient in proliferation and fail to undergo cell cycle progression or to express genes controlling cell cycle progression. In addition, the mice lack NK cells, develop splenomegaly, and have T cells with an activated phenotype, phenotypes seen in IL-2 receptor beta chain-deficient mice. These phenotypes are not seen in mice lacking Stat5a or Stat5b alone. The results demonstrate that the Stat5 proteins, redundantly, are essential mediators of IL-2 signaling in T cells.


Assuntos
Ciclo Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Interleucina-2/fisiologia , Proteínas do Leite , Linfócitos T/citologia , Transativadores/fisiologia , Animais , Divisão Celular , Células Cultivadas , Citometria de Fluxo , Camundongos , Camundongos Mutantes , Fator de Transcrição STAT5 , Timo/citologia , Timo/metabolismo
14.
Proc Natl Acad Sci U S A ; 96(1): 232-7, 1999 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-9874801

RESUMO

Human severe combined immunodeficiency (SCID) can be caused by defects in Janus kinase 3 (JAK3)-dependent cytokine signaling pathways. As a result, patients are at high risk of life-threatening infection. A JAK3 -/- SCID mouse model for the human disease has been used to test whether transplant with retrovirally transduced bone marrow (BM) cells (JAK3 BMT) could restore immunity to an influenza A virus. The immune responses also were compared directly with those for mice transplanted with wild-type BM (+/+ BMT). After infection, approximately 90% of the JAK3 BMT or +/+ BMT mice survived, whereas all of the JAK3 -/- mice died within 29 days. Normal levels of influenza-specific IgG were present in plasma from JAK3 BMT mice at 14 days after respiratory challenge, indicating restoration of B cell function. Influenza-specific CD4(+) and CD8(+) T cells were detected in the spleen and lymph nodes, and virus-specific CD8(+) effectors localized to the lungs of the JAK3 BMT mice. The kinetics of the specific host response correlated with complete clearance of the virus within 2 weeks of the initial exposure. By contrast, the JAK3 -/- mice did not show any evidence of viral immunity and were unable to control this viral pneumonia. Retroviral-mediated JAK3 gene transfer thus restores diverse aspects of cellular and humoral immunity and has obvious potential for human autologous BMT.


Assuntos
Terapia Genética/métodos , Vírus da Influenza A/imunologia , Proteínas Tirosina Quinases/genética , Imunodeficiência Combinada Severa/terapia , Animais , Anticorpos Antivirais/sangue , Transplante de Medula Óssea , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Imunidade Celular , Memória Imunológica , Janus Quinase 3 , Camundongos , Camundongos Mutantes , Imunodeficiência Combinada Severa/imunologia , Transformação Genética
15.
Blood ; 92(7): 2269-79, 1998 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9746764

RESUMO

Attempts to expand repopulating hematopoietic cells ex vivo have yielded only modest amplification in stem cell numbers. We now report that expression of an exogenous human multi-drug resistance 1 (MDR1) gene enables dramatic ex vivo stem cell expansion in the presence of early acting hematopoietic cytokines. Bone marrow cells were transduced with retroviral vectors expressing either the MDR1 gene or a variant of human dihydrofolate reductase (DHFR), and then expanded for 12 days in the presence of interleukin-3 (IL-3), IL-6, and stem cell factor. When these cells were injected into nonirradiated mice, high levels of long-term engraftment were only seen with MDR1-transduced grafts. To verify that expansion of MDR1-transduced repopulating cells had occurred, competitive repopulation assays were performed using MDR1 expanded grafts. These experiments showed progressive expansion of MDR1-transduced repopulating cells over the expansion period, with a 13-fold overall increase in stem cells after 12 days. In all of the experiments, mice transplanted with expanded MDR1-transduced stem cells developed a myeloproliferative disorder characterized by high peripheral white blood cell counts and splenomegaly. These results show that MDR1-transduced stem cells can be expanded in vitro using hematopoietic cytokines without any drug selection, but enforced stem cell self-renewal divisions can have adverse consequences.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transtornos Mieloproliferativos/etiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Divisão Celular , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/transplante , Genes MDR , Vetores Genéticos , Sobrevivência de Enxerto , Vírus do Sarcoma Murino de Harvey/genética , Hematopoese/efeitos dos fármacos , Humanos , Interleucina-3/farmacologia , Interleucina-6/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Fator de Células-Tronco/farmacologia , Transfecção
16.
Biochem Pharmacol ; 56(1): 31-9, 1998 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-9698086

RESUMO

Acrolein is a highly reactive and cytotoxic by-product released during activation of oxazaphosphorine (OAP) anticancer alkylating agents. Previously, we demonstrated that transfected human aldehyde dehydrogenase (ALDH, EC 1.2.1.3) isozymes (class 1 or 3) protect V79/SD1 cells from mafosfamide (MAF) cytotoxicity, but protection from 4-hydroperoxy-cyclophosphamide (4-hpCPA) was weaker. Acrolein, an ALDH inhibitor, may be detoxified by conjugation with the nucleophilic thiol 2-mercaptoethanesulfonate (MESNA), which is released from MAF but not from 4-hpCPA. We examined the effect of acrolein or acrolein/thiol conjugates on ALDH activity in vitro. We found that both ALDH isozymes were inhibited by acrolein, with IC50 values of 35 and 144 microM for ALDH-1 or ALDH-3, respectively. Both isozymes were partially protected by NAD+ cofactor, being at least five-fold more sensitive to acrolein if added before cofactor. In contrast, thiol conjugates of acrolein did not inhibit ALDH-3 activity, but were substrates only for ALDH-1. Further, acrolein was shown to be oxidized by ALDH-3, but not by ALDH-1. The effect of acrolein on ALDH-mediated resistance to OAP agents in intact cells was also examined. In control cells (without ALDH expression), acrolein and 4-hpCPA rapidly depleted intracellular GSH levels, whereas the effect of MAF was much less. Depletion of GSH by preincubation of V79/SD1 cells with a low concentration of acrolein (2 microM) before MAF exposure caused a two-fold reduction in ALDH-mediated resistance. Conversely, protection from 4-hpCPA cytotoxicity was enhanced by the addition of thiols (GSH, 2-mercaptoethanesulfonate, or N-acetylcysteine) during the drug exposure. These results suggest 1) that thiol content is an important determinant of the OAP resistance conferred by ALDH isoenzymes; and 2) a new mechanism whereby thiol modulation could increase the therapeutic index of OAP chemotherapy.


Assuntos
Acroleína/farmacologia , Aldeído Desidrogenase/metabolismo , Antineoplásicos Alquilantes/farmacologia , Isoenzimas/metabolismo , Compostos de Sulfidrila/metabolismo , Acroleína/metabolismo , Aldeído Desidrogenase/antagonistas & inibidores , Animais , Linhagem Celular , Cricetinae , Cricetulus , Glutationa/metabolismo , Humanos , Isoenzimas/antagonistas & inibidores
17.
Infect Immun ; 66(8): 3643-8, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9673244

RESUMO

We have developed a novel epitope delivery system based on the insertion of peptides within a permissive loop of a bacterial superoxide dismutase molecule. This system allowed high-level expression of heterologous peptides in two mycobacterial vaccine strains, Mycobacterium bovis bacille Calmette-Guérin (BCG) and Mycobacterium vaccae. The broader application of the system was analyzed by preparation of constructs containing peptide epitopes from a range of infectious agents and allergens. We report detailed characterization of the immunogenicity of one such construct, in which an epitope from the Der p1 house dust mite allergen was expressed in M. vaccae. The construct was able to stimulate T-cell hybridomas specific for Der p1, and it induced peptide-specific gamma interferon responses when used to immunize naive mice. This novel expression system demonstrates new possibilities for the use of mycobacteria as vaccine delivery vehicles.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Epitopos de Linfócito T/imunologia , Glicoproteínas/imunologia , Mycobacterium bovis/imunologia , Mycobacterium/imunologia , Superóxido Dismutase/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Bactérias/genética , Antígenos de Dermatophagoides , Proteínas de Bactérias/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Clonagem Molecular , Feminino , Expressão Gênica , Interferon gama/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mycobacterium/genética , Mycobacterium bovis/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Recombinação Genética , Superóxido Dismutase/genética
18.
Eur J Biochem ; 251(3): 795-803, 1998 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9490054

RESUMO

We have refined the X-ray structures of two site-directed mutants of the iron-dependent superoxide dismutase (SOD) from Mycobacterium tuberculosis. These mutations which affect residue 145 in the enzyme (H145Q and H145E) were designed to alter its metal-ion specificity. This residue is either Gln or His in homologous SOD enzymes and has previously been shown to play a role in active-site interactions since its side-chain helps to coordinate the metal ion via a solvent molecule which is thought to be a hydroxide ion. The mutations were based on the observation that in the closely homologous manganese dependent SOD from Mycobacterium leprae, the only significant difference from the M. tuberculosis SOD within 10 A of the metal-binding site is the substitution of Gln for His at position 145. Hence an H145Q mutant of the M. tuberculosis (TB) SOD was engineered to investigate this residue's role in metal ion dependence and an isosteric H145E mutant was also expressed. The X-ray structures of the H145Q and H145E mutants have been solved at resolutions of 4.0 A and 2.5 A, respectively, confirming that neither mutation has any gross effects on the conformation of the enzyme or the structure of the active site. The residue substitutions are accommodated in the enzyme's three-dimensional structure by small local conformational changes. Peroxide inhibition experiments and atomic absorption spectroscopy establish surprisingly the H145E mutant SOD has manganese bound to it whereas the H145Q mutant SOD retains iron as the active-site metal. This alteration in metal specificity may reflect on the preference of manganese ions for anionic ligands.


Assuntos
Ferro/metabolismo , Mycobacterium tuberculosis/enzimologia , Conformação Proteica , Superóxido Dismutase/química , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X/métodos , Ferro/farmacologia , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mycobacterium/enzimologia , Mutação Puntual , Reação em Cadeia da Polimerase , Estrutura Terciária de Proteína , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Superóxido Dismutase/biossíntese
19.
Nat Med ; 4(1): 58-64, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9427607

RESUMO

Janus kinase-3 (JAK3) deficiency has recently been identified as a cause of severe combined immunodeficiency (SCID) in humans. We used a mouse model of Jak3-deficient SCID to test a gene therapy approach for treatment of this disease. Transfer of a Jak3 retroviral vector to repopulating hematopoietic stem cells resulted in increased numbers of T and B lymphocytes, reversal of hypogammaglobulinemia, restoration of T-cell activation upon stimulation with mitogens, and development of an antigen-specific immune response after immunization. Analysis for vector copy number in lymphoid and myeloid populations showed a large in vivo selective advantage for Jak3-expressing lymphoid cells. These results show that gene replacement is a feasible treatment strategy for this disease and that naturally occurring in vivo selection of corrected cells is an important advantage of this approach.


Assuntos
Linfócitos B/imunologia , Terapia Genética/métodos , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/terapia , Linfócitos T/imunologia , Agamaglobulinemia/etiologia , Agamaglobulinemia/terapia , Animais , Formação de Anticorpos , Células da Medula Óssea/citologia , Transplante de Medula Óssea/métodos , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Janus Quinase 3 , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Reação em Cadeia da Polimerase , Proteínas Tirosina Quinases/biossíntese , Retroviridae , Imunodeficiência Combinada Severa/enzimologia , Baço/imunologia
20.
Hum Gene Ther ; 8(13): 1531-43, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9322086

RESUMO

Class 1 aldehyde dehydrogenases (ALDH-1) function as drug resistance gene products by catalyzing the irreversible conversion of aldophosphamide, an active metabolite of cyclophosphamide, to an inert compound. Because the dose-limiting toxicity of cyclophosphamide is myelosuppression, retrovirus-mediated transfer of ALDH-1 to bone marrow cells has been proposed as a protective strategy. Here we show that expression of ALDH-1 vectors was problematic due to low levels of ALDH-1 mRNA accumulation. A number of vectors containing several different ALDH-1 cDNAs were introduced into a variety of different cell lines either by transfection or transduction. Detectable ALDH-1 protein and enzyme activity was only seen in one transfected cell clone. Cells transduced with ALDH-1 retroviral vectors had no detectable protein expression and very low levels of ALDH-1 mRNA. Analogous vectors containing other drug resistance cDNAs led to much higher levels of steady-state mRNA. The mRNA half-life from ALDH-1 vectors was less than 2 hr suggesting that vector-derived mRNAs were destabilized by ALDH-1 coding sequences. These results suggest that methods which increase the stability of ALDH-1 mRNAs will be important for increased drug resistance in retrovirally transduced hematopoietic cells.


Assuntos
Aldeído Desidrogenase/genética , Vetores Genéticos , RNA Mensageiro/genética , Retroviridae/genética , Animais , DNA Complementar , Resistência a Medicamentos/genética , Meia-Vida , Camundongos , RNA Mensageiro/metabolismo , Tetra-Hidrofolato Desidrogenase/genética , Transdução Genética , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA