Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cell Reports ; 18(7): 1436-1450, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37352852

RESUMEN

Over the last several years, a method has emerged that endows adult hepatocytes with in vitro proliferative capacity, producing chemically induced liver progenitors (CLiPs). However, there is a growing controversy regarding the origin of these cells. Here, we provide lineage tracing-based evidence that adult hepatocytes acquire proliferative capacity in vitro using rat and mouse models. Unexpectedly, we also found that the CLiP method allows biliary epithelial cells to acquire extensive proliferative capacity. Interestingly, after long-term culture, hepatocyte-derived cells (hepCLiPs) and biliary epithelial cell-derived cells (bilCLiPs) become similar in their gene expression patterns, and they both exhibit differentiation capacity to form hepatocyte-like cells. Finally, we provide evidence that hepCLiPs can repopulate injured mouse livers, reinforcing our earlier argument that CLiPs can be a cell source for liver regenerative medicine. This study advances our understanding of the origin of CLiPs and motivates the application of this technique in liver regenerative medicine.


Asunto(s)
Hepatocitos , Células Madre , Ratones , Ratas , Animales , Células Madre/metabolismo , Hígado , Células Epiteliales/metabolismo , Diferenciación Celular , Proliferación Celular
2.
Nat Commun ; 12(1): 346, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436641

RESUMEN

Anti-PD-1 therapy is used as a front-line treatment for many cancers, but mechanistic insight into this therapy resistance is still lacking. Here we generate a humanized (Hu)-mouse melanoma model by injecting fetal liver-derived CD34+ cells and implanting autologous thymus in immune-deficient NOD-scid IL2Rγnull (NSG) mice. Reconstituted Hu-mice are challenged with HLA-matched melanomas and treated with anti-PD-1, which results in restricted tumor growth but not complete regression. Tumor RNA-seq, multiplexed imaging and immunohistology staining show high expression of chemokines, as well as recruitment of FOXP3+ Treg and mast cells, in selective tumor regions. Reduced HLA-class I expression and CD8+/Granz B+ T cells homeostasis are observed in tumor regions where FOXP3+ Treg and mast cells co-localize, with such features associated with resistance to anti-PD-1 treatment. Combining anti-PD-1 with sunitinib or imatinib results in the depletion of mast cells and complete regression of tumors. Our results thus implicate mast cell depletion for improving the efficacy of anti-PD-1 therapy.


Asunto(s)
Resistencia a Antineoplásicos , Linfocitos Infiltrantes de Tumor/inmunología , Mastocitos/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Mastocitos/efectos de los fármacos , Melanoma/inmunología , Melanoma/patología , Melanoma/terapia , Ratones Transgénicos , Receptor de Muerte Celular Programada 1/metabolismo , Sunitinib/farmacología , Sunitinib/uso terapéutico , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
3.
Comp Med ; 68(4): 261-268, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29898804

RESUMEN

Immunocompromised mice are used frequently in biomedical research, in part because they accommodate the engraftment and study of primary human cells within a mouse model; however, these animals are susceptible to opportunistic infections and require special husbandry considerations. In 2015, an outbreak marked by high morbidity but low mortality swept through a colony of immunocompromised mice; this outbreak rapidly affected 75% of the colony and ultimately required complete depopulation of the barrier suite. Conventional microbiologic and molecular diagnostics were unsuccessful in determining the cause; therefore, we explored culture-independent methods to broadly profile the microbial community in the feces of affected animals. This approach identified 4 bacterial taxa- Candidatus Arthromitus, Clostridium celatum, Clostridiales bacterium VE202-01, and Bifidobacterium pseudolongum strain PV8-2- that were significantly enriched in the affected mice. Based on these results, specific changes were made to the animal husbandry procedures for immunocompromised mice. This case report highlights the utility of culture-independent methods in laboratory animal diagnostics.


Asunto(s)
Diarrea/microbiología , Heces/microbiología , Microbiota/genética , Animales , Diarrea/epidemiología , Brotes de Enfermedades , Variación Genética , Huésped Inmunocomprometido , Metagenómica , Ratones , Ratones Endogámicos NOD , Ratones SCID , ARN Ribosómico 16S/química , Análisis de Secuencia de ARN , Secuenciación Completa del Genoma
4.
Haematologica ; 103(6): 959-971, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29545344

RESUMEN

Patient-derived xenotransplantation models of human myeloid diseases including acute myeloid leukemia, myelodysplastic syndromes and myeloproliferative neoplasms are essential for studying the biology of the diseases in pre-clinical studies. However, few studies have used these models for comparative purposes. Previous work has shown that acute myeloid leukemia blasts respond to human hematopoietic cytokines whereas myelodysplastic syndrome cells do not. We compared the engraftment of acute myeloid leukemia cells and myelodysplastic syndrome cells in NSG mice to that in NSG-S mice, which have transgene expression of human cytokines. We observed that only 50% of all primary acute myeloid leukemia samples (n=77) transplanted in NSG mice provided useful levels of engraftment (>0.5% human blasts in bone marrow). In contrast, 82% of primary acute myeloid leukemia samples engrafted in NSG-S mice with higher leukemic burden and shortened survival. Additionally, all of 5 injected samples from patients with myelodysplastic syndrome showed persistent engraftment on week 6; however, engraftment was mostly low (<2%), did not increase over time, and was only transiently affected by the use of NSG-S mice. Co-injection of mesenchymal stem cells did not enhance human myelodysplastic syndrome cell engraftment. Overall, we conclude that engraftment of acute myeloid leukemia samples is more robust compared to that of myelodysplastic syndrome samples and unlike those, acute myeloid leukemia cells respond positively to human cytokines, whereas myelodysplastic syndrome cells demonstrate a general unresponsiveness to them.


Asunto(s)
Citocinas/metabolismo , Supervivencia de Injerto/inmunología , Huésped Inmunocomprometido , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Síndromes Mielodisplásicos/inmunología , Síndromes Mielodisplásicos/metabolismo , Animales , Trasplante de Médula Ósea , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Humanos , Leucemia Mieloide Aguda/terapia , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Síndromes Mielodisplásicos/terapia , Trasplante Heterólogo
5.
PLoS Pathog ; 13(10): e1006613, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29023549

RESUMEN

HIV is adept at avoiding naturally generated T cell responses; therefore, there is a need to develop HIV-specific T cells with greater potency for use in HIV cure strategies. Starting with a CD4-based chimeric antigen receptor (CAR) that was previously used without toxicity in clinical trials, we optimized the vector backbone, promoter, HIV targeting moiety, and transmembrane and signaling domains to determine which components augmented the ability of T cells to control HIV replication. This re-engineered CAR was at least 50-fold more potent in vitro at controlling HIV replication than the original CD4 CAR, or a TCR-based approach, and substantially better than broadly neutralizing antibody-based CARs. A humanized mouse model of HIV infection demonstrated that T cells expressing optimized CARs were superior at expanding in response to antigen, protecting CD4 T cells from infection, and reducing viral loads compared to T cells expressing the original, clinical trial CAR. Moreover, in a humanized mouse model of HIV treatment, CD4 CAR T cells containing the 4-1BB costimulatory domain controlled HIV spread after ART removal better than analogous CAR T cells containing the CD28 costimulatory domain. Together, these data indicate that potent HIV-specific T cells can be generated using improved CAR design and that CAR T cells could be important components of an HIV cure strategy.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/terapia , Infecciones por VIH/virología , VIH-1/fisiología , Recoverina/inmunología , Replicación Viral , Anticuerpos Neutralizantes/inmunología , Infecciones por VIH/inmunología , Humanos , Transducción de Señal/fisiología
6.
Nat Commun ; 8: 14630, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28251988

RESUMEN

Monoclonal antibodies are one of the fastest growing classes of pharmaceutical products, however, their potential is limited by the high cost of development and manufacturing. Here we present a safe and cost-effective platform for in vivo expression of therapeutic antibodies using nucleoside-modified mRNA. To demonstrate feasibility and protective efficacy, nucleoside-modified mRNAs encoding the light and heavy chains of the broadly neutralizing anti-HIV-1 antibody VRC01 are generated and encapsulated into lipid nanoparticles. Systemic administration of 1.4 mg kg-1 of mRNA into mice results in ∼170 µg ml-1 VRC01 antibody concentrations in the plasma 24 h post injection. Weekly injections of 1 mg kg-1 of mRNA into immunodeficient mice maintain trough VRC01 levels above 40 µg ml-1. Most importantly, the translated antibody from a single injection of VRC01 mRNA protects humanized mice from intravenous HIV-1 challenge, demonstrating that nucleoside-modified mRNA represents a viable delivery platform for passive immunotherapy against HIV-1 with expansion to a variety of diseases.


Asunto(s)
Anticuerpos Neutralizantes/genética , VIH-1/efectos de los fármacos , Nucleósidos/química , ARN Mensajero/administración & dosificación , Animales , Anticuerpos Monoclonales/genética , Anticuerpos ampliamente neutralizantes , Esquema de Medicación , Femenino , Anticuerpos Anti-VIH/biosíntesis , Infecciones por VIH/inmunología , Infecciones por VIH/terapia , VIH-1/inmunología , Humanos , Inmunización Pasiva , Lípidos/química , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Nanopartículas/química , ARN Mensajero/química , ARN Mensajero/farmacología , ARN Mensajero/uso terapéutico
7.
PLoS Pathog ; 12(11): e1005983, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27855210

RESUMEN

HIV-1 entry can be inhibited by soluble peptides from the gp41 heptad repeat-2 (HR2) domain that interfere with formation of the 6-helix bundle during fusion. Inhibition has also been seen when these peptides are conjugated to anchoring molecules and over-expressed on the cell surface. We hypothesized that potent anti-HIV activity could be achieved if a 34 amino acid peptide from HR2 (C34) were brought to the site of virus-cell interactions by conjugation to the amino termini of HIV-1 coreceptors CCR5 or CXCR4. C34-conjugated coreceptors were expressed on the surface of T cell lines and primary CD4 T cells, retained the ability to mediate chemotaxis in response to cognate chemokines, and were highly resistant to HIV-1 utilization for entry. Notably, C34-conjugated CCR5 and CXCR4 each exhibited potent and broad inhibition of HIV-1 isolates from diverse clades irrespective of tropism (i.e., each could inhibit R5, X4 and dual-tropic isolates). This inhibition was highly specific and dependent on positioning of the peptide, as HIV-1 infection was poorly inhibited when C34 was conjugated to the amino terminus of CD4. C34-conjugated coreceptors could also inhibit HIV-1 isolates that were resistant to the soluble HR2 peptide inhibitor, enfuvirtide. When introduced into primary cells, CD4 T cells expressing C34-conjugated coreceptors exhibited physiologic responses to T cell activation while inhibiting diverse HIV-1 isolates, and cells containing C34-conjugated CXCR4 expanded during HIV-1 infection in vitro and in a humanized mouse model. Notably, the C34-conjugated peptide exerted greater HIV-1 inhibition when conjugated to CXCR4 than to CCR5. Thus, antiviral effects of HR2 peptides can be specifically directed to the site of viral entry where they provide potent and broad inhibition of HIV-1. This approach to engineer HIV-1 resistance in functional CD4 T cells may provide a novel cell-based therapeutic for controlling HIV infection in humans.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Proteína gp41 de Envoltorio del VIH/metabolismo , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Fragmentos de Péptidos/metabolismo , Receptores CXCR4/metabolismo , Internalización del Virus , Animales , Linfocitos T CD4-Positivos/metabolismo , Citometría de Flujo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos NOD
8.
Blood ; 123(1): 61-9, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24162716

RESUMEN

HIV-1 entry into CD4(+) T cells requires binding of the virus to CD4 followed by engagement of either the C-C chemokine receptor 5 (CCR5) or C-X-C chemokine receptor 4 (CXCR4) coreceptor. Pharmacologic blockade or genetic inactivation of either coreceptor protects cells from infection by viruses that exclusively use the targeted coreceptor. We have used zinc-finger nucleases to drive the simultaneous genetic modification of both ccr5 and cxcr4 in primary human CD4(+) T cells. These gene-modified cells proliferated normally and were resistant to both CCR5- and CXCR4-using HIV-1 in vitro. When introduced into a humanized mouse model of HIV-1 infection, these coreceptor negative cells engraft and traffic normally, and are protected from infection with CCR5- and CXCR4-using HIV-1 strains. These data suggest that simultaneous disruption of the HIV coreceptors may provide a useful approach for the long-term, drug-free treatment of established HIV-1 infections.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Endodesoxirribonucleasas/metabolismo , Infecciones por VIH/inmunología , Receptores CCR5/genética , Receptores CXCR4/genética , Dedos de Zinc , Animales , Linfocitos T CD4-Positivos/citología , Proliferación Celular , Femenino , Células HEK293 , Infecciones por VIH/prevención & control , Infecciones por VIH/terapia , VIH-1 , Humanos , Masculino , Ratones , Receptores de Quimiocina/metabolismo
9.
PLoS Pathog ; 7(4): e1002020, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21533216

RESUMEN

HIV-1 entry requires the cell surface expression of CD4 and either the CCR5 or CXCR4 coreceptors on host cells. Individuals homozygous for the ccr5Δ32 polymorphism do not express CCR5 and are protected from infection by CCR5-tropic (R5) virus strains. As an approach to inactivating CCR5, we introduced CCR5-specific zinc-finger nucleases into human CD4+ T cells prior to adoptive transfer, but the need to protect cells from virus strains that use CXCR4 (X4) in place of or in addition to CCR5 (R5X4) remains. Here we describe engineering a pair of zinc finger nucleases that, when introduced into human T cells, efficiently disrupt cxcr4 by cleavage and error-prone non-homologous DNA end-joining. The resulting cells proliferated normally and were resistant to infection by X4-tropic HIV-1 strains. CXCR4 could also be inactivated in ccr5Δ32 CD4+ T cells, and we show that such cells were resistant to all strains of HIV-1 tested. Loss of CXCR4 also provided protection from X4 HIV-1 in a humanized mouse model, though this protection was lost over time due to the emergence of R5-tropic viral mutants. These data suggest that CXCR4-specific ZFNs may prove useful in establishing resistance to CXCR4-tropic HIV for autologous transplant in HIV-infected individuals.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Desoxirribonucleasas/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Receptores CXCR4/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/trasplante , Proliferación Celular , Desoxirribonucleasas/biosíntesis , Desoxirribonucleasas/genética , Modelos Animales de Enfermedad , Ingeniería Genética , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , Infecciones por VIH/terapia , VIH-1/genética , VIH-1/metabolismo , Humanos , Macaca mulatta , Ratones , Receptores CCR5/genética , Receptores CCR5/inmunología , Receptores CCR5/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Trasplante Autólogo , Trasplante Heterólogo , Internalización del Virus
10.
J Clin Invest ; 121(1): 384-95, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21157036

RESUMEN

Human leukemic stem cells, like other cancer stem cells, are hypothesized to be rare, capable of incomplete differentiation, and restricted to a phenotype associated with early hematopoietic progenitors or stem cells. However, recent work in other types of tumors has challenged the cancer stem cell model. Using a robust model of xenotransplantation based on NOD/SCID/IL2Rγc-deficient mice, we confirmed that human leukemic stem cells, functionally defined by us as SCID leukemia-initiating cells (SL-ICs), are rare in acute myelogenous leukemia (AML). In contrast to previous results, SL-ICs were found among cells expressing lineage markers (i.e., among Lin+ cells), CD38, or CD45RA, all markers associated with normal committed progenitors. Remarkably, each engrafting fraction consistently recapitulated the original phenotypic diversity of the primary AML specimen and contained self-renewing leukemic stem cells, as demonstrated by secondary transplants. While SL-ICs were enriched in the Lin-CD38- fraction compared with the other fractions analyzed, SL-ICs in this fraction represented only one-third of all SL-ICs present in the unfractionated specimen. These results indicate that human AML stem cells are rare and enriched but not restricted to the phenotype associated with normal primitive hematopoietic cells. These results suggest a plasticity of the cancer stem cell phenotype that we believe has not been previously described.


Asunto(s)
Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/trasplante , Animales , Secuencia de Bases , Diferenciación Celular , Linaje de la Célula , Cartilla de ADN/genética , Femenino , Células Madre Hematopoyéticas/patología , Humanos , Inmunofenotipificación , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/fisiopatología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Modelos Biológicos , Células Madre Neoplásicas/fisiología , Trasplante Heterólogo
11.
Atherosclerosis ; 181(2): 271-4, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16039280

RESUMEN

Endothelial lipase (EL), a new member of the triacylglycerol lipase gene family, is a key enzyme in HDL metabolism. The EL expression pattern in humans was reported to be unique and complementary to that documented for lipoprotein lipase. The regulatory elements responsible for the tissue-specific EL expression are not identified yet. In order to confine these sequences to a defined region of the EL promoter, we analyzed EL mRNA expression in human EL transgenic mice expressing EL under the control of the endogenous human promoter. We identified small intestine, mammary gland, adipose tissue and the adrenal gland as previously unknown tissues to express EL. Our data demonstrate that regulatory elements within 11.4 kb of 5' and 9.9 kb of 3' human EL flanking region promote the expression of EL in small intestine, ovary, testis, mammary gland, brain, lung, aorta, adipose tissue and the adrenals, whereas regulatory sequences located between 27.4 and 11.4 kb of 5' or 9.9 and 48.7 kb of 3' human EL flanking region seem to be responsible for kidney-specific EL expression.


Asunto(s)
Regulación de la Expresión Génica , Lipasa/genética , Regiones Promotoras Genéticas/fisiología , Regiones no Traducidas 3'/genética , Regiones no Traducidas 5'/genética , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos , Embarazo , ARN Mensajero/análisis
12.
Arterioscler Thromb Vasc Biol ; 23(10): 1914-20, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12933536

RESUMEN

OBJECTIVE: An inverse and independent association between plasma levels of apolipoprotein (apo) A-I and coronary heart disease in humans is well established. ApoA-I is the primary protein component of HDL and is thought to play an important role in mediating several of the atheroprotective effects of HDL. However, studies of the effects of apoA-I deficiency on the development of atherosclerosis in mice have not been definitive. We examined the effects of apoA-I deficiency on plasma lipids and atherosclerosis in LDL receptor-deficient mice fed a chow diet for up to 22 months. METHODS AND RESULTS: Both apoA-I-deficient (apoA-I-/-)/LDL receptor-deficient (LDLR-/-) and LDLR-/- mice had a similar moderate elevation of non-HDL cholesterol (non-HDL-C). Unlike previous studies of apoA-I deficiency in which the HDL-C levels were extremely low, the apoA-I-/-/LDLR-/- mice in this study had substantial levels of HDL-C that were similar to wild-type mice. Despite similar levels of non-HDL-C and substantial levels of HDL-C, apoA-I-/-/LDLR-/- mice develop significantly more atherosclerosis (up to a 5-fold increase) and oxidant stress (39% increase) than LDLR-/- mice. CONCLUSIONS: These results demonstrate that despite normal levels of HDL-C, apoA-I deficiency is associated with a significant loss of protection from the formation of atherosclerosis in LDLR-/- mice fed a chow diet.


Asunto(s)
Apolipoproteína A-I/deficiencia , Arteriosclerosis/etiología , HDL-Colesterol/sangre , Receptores de LDL/deficiencia , Animales , Apolipoproteína A-I/genética , LDL-Colesterol/sangre , VLDL-Colesterol/sangre , Dieta , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Receptores de LDL/genética , Triglicéridos/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...