Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
2.
Blood Adv ; 8(3): 513-522, 2024 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-37871306

RESUMEN

ABSTRACT: Outcomes in patients with relapsed diffuse large B-cell lymphoma (DLBCL) who undergo autologous stem cell transplant (auto-SCT) are poor. Blinatumomab is a CD3/CD19 bispecific T-cell engager that directs cytotoxic T cells to CD19+ cells. Here, we performed a pilot study of blinatumomab consolidation after auto-SCT for 14 patients with DLBCL or transformed follicular lymphoma. All patients underwent standard-of-care auto-SCT with carmustine, etoposide, cytarabine, and melphalan (BEAM) conditioning followed by 1 cycle (4 weeks continuous infusion) of blinatumomab consolidation starting at day 42 after auto-SCT. All 14 patients treated on study completed BEAM auto-SCT and 1 cycle of posttransplant blinatumomab. Five patients developed grade 1 cytokine release syndrome (CRS), with no grade 2 or higher CRS. Immune effector cell-associated neurotoxicity syndrome was not observed. Patients were followed up for 3 years after auto-SCT, with median follow-up of 37 (range, 12-65) months. One-hundred days after auto-SCT (1 month after blinatumomab consolidation), 12 patients (86%) had achieved complete remission. At 1 year after auto-SCT, 7 patients (50%) remained in CR, and 1 patient had died of progressive disease. Patients who relapsed had a lower CD8:CD4 T-cell ratio before starting blinatumomab than patients who remained in remission. This pilot study demonstrates blinatumomab consolidation after auto-SCT is safe and well tolerated. Strategies to increase the CD8:CD4 ratio and use additional cycles of consolidation in a larger randomized trial are needed to confirm the efficacy of consolidation with blinatumomab after auto-SCT. This trial was registered at www.clinicaltrials.gov as #NCT03072771.


Asunto(s)
Anticuerpos Biespecíficos , Trasplante de Células Madre Hematopoyéticas , Linfoma de Células B Grandes Difuso , Linfoma no Hodgkin , Humanos , Proyectos Piloto , Inducción de Remisión , Trasplante Autólogo , Recurrencia Local de Neoplasia , Trasplante de Células Madre
3.
Open Forum Infect Dis ; 10(11): ofad518, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37953814

RESUMEN

Background: Despite improvements in prevention and treatment, severe coronavirus disease 2019 (COVID-19) is associated with high mortality. Phosphoinositide 3-kinase (PI3K) pathways contribute to cytokine and cell-mediated lung inflammation. We conducted a randomized, placebo-controlled, double-blind pilot trial to determine the feasibility, safety, and preliminary activity of duvelisib, a PI3Kδγ inhibitor, for the treatment of COVID-19 critical illness. Methods: We enrolled adults aged ≥18 years with a primary diagnosis of COVID-19 with hypoxic respiratory failure, shock, and/or new cardiac disease, without improvement after at least 48 hours of corticosteroid. Participants received duvelisib (25 mg) or placebo for up to 10 days. Participants had daily semi-quantitative viral load measurements performed. Dose modifications were protocol driven due to adverse events (AEs) or logarithmic change in viral load. The primary endpoint was 28-day overall survival (OS). Secondary endpoints included hospital and intensive care unit length of stay, 60-day OS, and duration of critical care interventions. Safety endpoints included viral kinetics and AEs. Exploratory endpoints included serial cytokine measurements and cytometric analysis. Results: Fifteen patients were treated in the duvelisib cohort, and 13 in the placebo cohort. OS at 28 days was 67% (95% confidence interval [CI], 38%-88%) compared to 62% (95% CI, 32%-86%) for placebo (P = .544). Sixty-day OS was 60% versus 46%, respectively (hazard ratio, 0.66 [95% CI, .22-1.96]; P = .454). Other secondary outcomes were comparable. Duvelisib was associated with lower inflammatory cytokines. Conclusions: In this pilot study, duvelisib did not significantly improve 28-day OS compared to placebo for severe COVID-19. Duvelisib appeared safe in this critically ill population and was associated with reduction in cytokines implicated in COVID-19 and acute respiratory distress syndrome, supporting further investigation. Clinical Trials Registration: NCT04372602.

4.
Soft Matter ; 19(43): 8329-8336, 2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-37869971

RESUMEN

Direct ink writing, a versatile method of 3D and 4D printing, requires the precise placement of a nozzle just above the print surface to prevent fluid instabilities that cause deviations from the prescribed print path. But what if one could harness the instability associated with the spontaneously folding or coiling of a thin stream of viscous fluid, i.e. use the "fluid rope trick" to write specified patterns on a substrate? Here we use Deep Reinforcement Learning to derive control strategies for the motion of the extruding nozzle and thus the fluid patterns that are deposited on the surface. The method proceeds by having a learner (nozzle) repeatedly interact with the environment (a viscous filament simulator), and improves its strategy using the results of this experience. We demonstrate the outcome of the learned control instructions using experiments to manipulate a falling viscous jet and create cursive writing patterns and Pollockian paintings on substrates.

5.
Blood ; 141(14): 1718-1723, 2023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-36563336

RESUMEN

Acute myeloid leukemia (AML) relapse is one of the most common and significant adverse events following allogeneic hematopoietic cell transplantation (HCT). Downregulation of major histocompatibility class II (MHC-II) surface expression on AML blasts may represent a mechanism of escape from the graft-versus-malignancy effect and facilitate relapse. We hypothesized that T-cell immunotherapies targeting AML antigens would upregulate MHC-II surface expression via localized release of interferon gamma (IFN-γ), a protein known to upregulate MHC-II expression via JAK-STAT signaling. We demonstrate that flotetuzumab (FLZ), a CD123 × CD3 bispecific DART molecule, and chimeric antigen receptor expressing T cells targeting CD123, CD33, or CD371 upregulate MHC-II surface expression in vitro on a THP-1 AML cell line with intermediate MHC-II expression and 4 primary AML samples from patients relapsing after HCT with low MHC-II expression. We additionally show that FLZ upregulates MHC-II expression in a patient-derived xenograft model and in patients with relapsed or refractory AML who were treated with FLZ in a clinical trial. Finally, we report that FLZ-induced MHC-II upregulation is mediated by IFN-γ. In conclusion, we provide evidence that T-cell immunotherapies targeting relapsed AML can kill AML via both MHC-independent mechanisms and by an MHC-dependent mechanism through local release of IFN-γ and subsequent upregulation of MHC-II expression.


Asunto(s)
Anticuerpos Biespecíficos , Antineoplásicos , Leucemia Mieloide Aguda , Humanos , Linfocitos T , Subunidad alfa del Receptor de Interleucina-3 , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Interferón gamma , Complejo CD3 , Inmunoterapia , Recurrencia
6.
Leuk Lymphoma ; 62(6): 1441-1449, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33467957

RESUMEN

Interactions between the bone marrow microenvironment and MDS tumor clones play a role in pathogenesis and response to treatment. We hypothesized G-CSF and plerixafor may enhance sensitivity to azacitidine in MDS. Twenty-eight patients with MDS were treated with plerixafor, G-CSF and azacitidine with a standard 3 + 3 design. Subjects received G-CSF 10 mcg/kg D1-D8, plerixafor D4-D8, and azacitidine 75 mg/m2 D4-D8, but the trial was amended to reduce G-CSF dose to 5 mcg/kg for 5 days after 2 patients had significant leukocytosis. Plerixafor was dose escalated to 560 mcg/kg/day without dose limiting toxicity. Two complete responses and 6 marrow responses were seen for an overall response rate (ORR) of 36% in evaluable patients, and ORR of 53% in patients receiving the triplet. Evidence of mobilization correlated with a higher ORR, 60% vs. 17%. Plerixafor, G-CSF and azacitidine appears tolerable when given over 5 days and has encouraging response rates.KEY POINTSPlerixafor and G-CSF can be safely combined with azacitidine for 5 days in patients with MDS.The overall response rate of 53% for evaluable patients with this regimen is higher than expected and more responses were seen in patients with blast mobilization.


Asunto(s)
Compuestos Heterocíclicos , Síndromes Mielodisplásicos , Azacitidina/efectos adversos , Bencilaminas , Ciclamas , Factor Estimulante de Colonias de Granulocitos , Movilización de Célula Madre Hematopoyética , Compuestos Heterocíclicos/efectos adversos , Humanos , Síndromes Mielodisplásicos/tratamiento farmacológico
7.
J Clin Invest ; 129(7): 2745-2759, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31085833

RESUMEN

Mobilized peripheral blood has become the primary source of hematopoietic stem and progenitor cells (HSPCs) for stem cell transplantation, with a five-day course of granulocyte colony stimulating factor (G-CSF) as the most common regimen used for HSPC mobilization. The CXCR4 inhibitor, plerixafor, is a more rapid mobilizer, yet not potent enough when used as a single agent, thus emphasizing the need for faster acting agents with more predictable mobilization responses and fewer side effects. We sought to improve hematopoietic stem cell transplantation by developing a new mobilization strategy in mice through combined targeting of the chemokine receptor CXCR2 and the very late antigen 4 (VLA4) integrin. Rapid and synergistic mobilization of HSPCs along with an enhanced recruitment of true HSCs was achieved when a CXCR2 agonist was co-administered in conjunction with a VLA4 inhibitor. Mechanistic studies revealed involvement of CXCR2 expressed on BM stroma in addition to stimulation of the receptor on granulocytes in the regulation of HSPC localization and egress. Given the rapid kinetics and potency of HSPC mobilization provided by the VLA4 inhibitor and CXCR2 agonist combination in mice compared to currently approved HSPC mobilization methods, it represents an exciting potential strategy for clinical development in the future.


Asunto(s)
Médula Ósea/metabolismo , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Integrina alfa4beta1 , Receptores de Interleucina-8B , Aloinjertos , Animales , Granulocitos/metabolismo , Integrina alfa4beta1/antagonistas & inhibidores , Integrina alfa4beta1/genética , Integrina alfa4beta1/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptores de Interleucina-8B/antagonistas & inhibidores , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo
8.
Blood ; 129(21): 2939-2949, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28400375

RESUMEN

Interaction between the chemokine receptor CXCR4 and its chief ligand CXCL12 plays a critical role in the retention and migration of hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM) microenvironment. In this study, qualitative and quantitative effects of long-term pharmacologic inhibition of the CXCR4/CXCL12 axis on the HSPC compartment were investigated by using 3 structurally unrelated small molecule CXCR4 antagonists. A >10-fold increase in mobilization efficiency was achieved by administering the antagonists as a subcutaneous continuous infusion for 2 weeks compared to a single bolus injection. A concurrent increase in self-renewing proliferation leading to a twofold to fourfold expansion of the HSPC pool in the BM was observed. The expanded BM showed a distinct repopulating advantage when tested in serial competitive transplantation experiments. Furthermore, major changes within the HSPC niche associated with previously described HSPC expansion strategies were not detected in bones treated with a CXCR4 antagonist infusion. Our data suggest that prolonged but reversible pharmacologic blockade of the CXCR4/CXCL12 axis represents an approach that releases HSPC with efficiency superior to any other known mobilization strategy and may also serve as an effective method to expand the BM HSPC pool.


Asunto(s)
Movilización de Célula Madre Hematopoyética/métodos , Células Madre Hematopoyéticas/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Nicho de Células Madre/efectos de los fármacos , Animales , Médula Ósea/metabolismo , Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Ratones , Ratones Transgénicos , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
9.
Blood ; 129(19): 2680-2692, 2017 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-28292947

RESUMEN

A single subcutaneous (SC) injection of plerixafor results in rapid mobilization of hematopoietic progenitors, but fails to mobilize 33% of normal allogeneic sibling donors in 1 apheresis. We hypothesized that changing the route of administration of plerixafor from SC to IV may overcome the low stem cell yields and allow collection in 1 day. A phase 1 trial followed by a phase 2 efficacy trial was conducted in allogeneic sibling donors. The optimal dose of IV plerixafor was determined to be 0.32 mg/kg. The primary outcome of reducing the failure to collect ≥2 × 106 CD34+/kg recipient weight in 1 apheresis collection to ≤10% was not reached. The failure rate was 34%. Studies evaluating the stem cell phenotype and gene expression revealed a novel plasmacytoid dendritic cell precursor preferentially mobilized by plerixafor with high interferon-α producing ability. The observed cytomegalovirus (CMV) viremia rate for patients at risk was low (15%), as were the rates of acute grade 2-4 graft-versus-host disease (GVHD) (21%). Day 100 treatment related mortality was low (3%). In conclusion, plerixafor results in rapid stem cell mobilization regardless of route of administration and resulted in novel cellular composition of the graft and favorable recipient outcomes. These trials were registered at clinicaltrials.gov as #NCT00241358 and #NCT00914849.


Asunto(s)
Movilización de Célula Madre Hematopoyética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Compuestos Heterocíclicos/farmacología , Células Madre de Sangre Periférica/efectos de los fármacos , Administración Intravenosa , Adulto , Anciano , Antígenos CD34/análisis , Bencilaminas , Eliminación de Componentes Sanguíneos , Ciclamas , Femenino , Enfermedad Injerto contra Huésped/etiología , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/farmacología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Compuestos Heterocíclicos/administración & dosificación , Compuestos Heterocíclicos/farmacocinética , Humanos , Masculino , Persona de Mediana Edad , Células Madre de Sangre Periférica/citología , Donantes de Tejidos , Transcriptoma/efectos de los fármacos , Trasplante Homólogo/efectos adversos , Trasplante Homólogo/métodos
10.
J Cell Sci ; 119(Pt 14): 2975-84, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16803873

RESUMEN

Thyroid hormone (TH or T3) and TH-receptor beta (TRbeta) have been reported to be relevant for cochlear development and hearing function. Mutations in the TRbeta gene result in deafness associated with resistance to TH syndrome. The effect of TRalpha1 on neither hearing function nor cochlear T3 target genes has been described to date. It is also uncertain whether TRalpha1 and TRbeta can act simultaneously on different target genes within a single cell. We focused on two concomitantly expressed outer hair cell genes, the potassium channel Kcnq4 and the motor protein prestin Slc26a5. In outer hair cells, TH enhanced the expression of the prestin gene through TRbeta. Simultaneously Kcnq4 expression was activated in the same cells by derepression of TRalpha1 aporeceptors mediated by an identified THresponse element, which modulates KCNQ4 promoter activity. We show that T3 target genes can differ in their sensitivity to TH receptors having the ligand either bound (holoreceptors) or not bound (aporeceptors) within single cells, and suggest a role for TRalpha1 in final cell differentiation.


Asunto(s)
Diferenciación Celular , Regulación de la Expresión Génica , Células Ciliadas Auditivas Externas/citología , Canales de Potasio KCNQ/genética , Proteínas/genética , Receptores alfa de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/metabolismo , Animales , Proteínas de Transporte de Anión , Secuencia de Bases , Células Cultivadas , Genes Dominantes/genética , Células Ciliadas Auditivas Externas/metabolismo , Humanos , Hipotiroidismo/metabolismo , Ratones , Datos de Secuencia Molecular , Mutación/genética , Regiones Promotoras Genéticas/genética , Ratas , Ratas Wistar , Elementos de Respuesta/genética , Transportadores de Sulfato , Receptores alfa de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/genética , Hormonas Tiroideas/deficiencia
11.
Growth Factors ; 23(1): 43-53, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16019426

RESUMEN

The connective tissue growth factor (CTGF) is a well-known fibroblast mitogen and angiogenic factor that plays an important role in bone formation during embryogenesis. In the adult, CTGF is involved in wound healing as well as fibrotic and vascular disease. However, little is known about its physiological functions under non-pathological conditions in the adult organism. Here, we describe the cellular site of the CTGF mRNA expression in adult male and female mice as revealed by in situ hybridization histochemistry. Strong and persistent CTGF gene expression was particularly prominent in the mesenchyme of the cardiovascular system (aorta, auricular tissue, renal glomeruli), the mesenchyme surrounding the ovarian follicles or the testicular tubes in the gonadal tissue, and the subcapsular mesenchyme bordering densely innervated parts of whisker hair vibrissae. CTGF hybridization signals were not observed in the mesenchyme of many other organs including gut, muscle, liver or most parts of the lymphatic tissue. Strong expression was also present in the primary (early) ovarian follicles, the epithelium of the deep uterine glands and on myenteric ganglia neurons. These data suggest a selective and continuous mesenchymal function in the gonads and those tissues attracting very strong vascular supply or peripheral innervation. CTGF may also be involved in the cyclical proliferation of the uterine gland epithelium and in the early stages of follicular maturation, as well as in the neuropeptide regulation in the gut, cardiovascular and renal systems.


Asunto(s)
Expresión Génica , Proteínas Inmediatas-Precoces/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Animales , Factor de Crecimiento del Tejido Conjuntivo , Femenino , Procesamiento de Imagen Asistido por Computador , Proteínas Inmediatas-Precoces/genética , Inmunohistoquímica , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Mesodermo/metabolismo , Ratones , Ratones Endogámicos , ARN Mensajero/metabolismo , Distribución Tisular
12.
Audiol Neurootol ; 9(2): 88-106, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14981357

RESUMEN

Pax8-/- mice do not develop thyroid follicular structures and thus provide an ideal animal model to study the consequences of congenital hypothyroidism. Despite their athyroidism, Pax8-/- mice survive up to postnatal day 21 (P21). No auditory brain stem responses (ABR) to sound could be recorded in these animals at 130 dB SPL, even at P21, when hearing reaches adult sensitivity in control mice. Abnormalities in the outer and middle ear structures were found in a considerable percentage of Pax8-/- animals. Maturation of the inner ear appeared delayed by about 1 week with respect to euthyroid controls. Hearing of adult Pax8-/- mice could be nearly normalized by early postnatal substitution with thyroxine (T(4)), but structural and functional restoration of hearing was incomplete. Even when T(4) substitution was initiated at P1, ABR thresholds, measured at 6 weeks of age or more, were increased by about 20 dB, and each day of delay in the start of T(4) substitution resulted in an additional threshold loss of about 4 dB. The most prominent structural deficit in Pax8-/- animals in which T(4) substitution was started at P8 or later was an abnormally thick tectorial membrane. In these late-substituted animals, disarray of stereovilli from inner and outer hair cells was observed and also outer hair cell loss was found, predominantly in the basal part of the cochlea. The degree of structural disorder increased the later T(4) substitution was initiated. The structural and functional consequences of postnatal athyroidism observed in Pax8-/- mice are largely in agreement with and extend those data obtained from hypothyroid animal models in which hypothyroidism was induced by goitrogenic agents (methimazole, propylthiouracil) or animal models with disrupted genes for the TSH receptor or the thyroid hormone receptors. The hearing loss and also the recovery effect by T(4) substitution in Pax8-/- mice is larger than that in the other models. Although Pax8-/- mice are born by euthyroid Pax8+/- dams, the Pax8-/- phenotype could not be completely restored by immediate postnatal T(4) substitution, indicating that some deficits are the consequence of prenatal T(4) deficiency of the offspring.


Asunto(s)
Cóclea/anomalías , Hipotiroidismo Congénito , Pérdida Auditiva/etiología , Proteínas Nucleares , Tiroxina/administración & dosificación , Animales , Umbral Auditivo , Cóclea/ultraestructura , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Oído Externo/anomalías , Oído Medio/anomalías , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Pérdida Auditiva/tratamiento farmacológico , Pérdida Auditiva/patología , Hipotiroidismo/complicaciones , Hipotiroidismo/tratamiento farmacológico , Masculino , Ratones , Ratones Noqueados , Microscopía Electrónica de Rastreo , Factor de Transcripción PAX8 , Factores de Transcripción Paired Box , Transactivadores/genética
13.
Endocrinology ; 145(3): 1276-83, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14617574

RESUMEN

Signaling mechanisms in pituitary morphogenesis as well as pituitary cell fate determination during early embryonic development are relatively well characterized. In contrast, the cues that determine the progression of the various anterior pituitary cell types during postnatal periods are poorly defined. Pax8-/- mice, which are born without a thyroid gland, were used to study the influence of thyroid hormones on the expression of pituitary hormones during early postnatal life. Serum pituitary hormones were determined by RIAs, and the pituitaries were analyzed by Northern blotting, in situ hybridization histochemistry, and immunocytochemistry. In 21-d-old Pax8-/- mice, the cellular composition of the anterior pituitary was dramatically distorted. Thyrotropes exhibited hypertrophy and hyperplasia, the number of detectable somatotropes was drastically reduced, and lactotropes were almost undetectable. Expression of LH and FSH was also reduced, but ACTH and proopiomelanocortin expression was not significantly different. Serum pituitary hormone levels were changed correspondingly. T(4) replacement therapy for variable time periods normalized TSH and GH mRNA expression within 3 d but not prolactin expression, not even when T(4) was administered for 6 d in combination with estradiol. These findings reveal the importance of thyroid hormones in developing the appropriate proportions of anterior pituitary cell types, especially with regard to lactotropes.


Asunto(s)
Proteínas de Unión al ADN/genética , Hipotiroidismo/fisiopatología , Proteínas Nucleares , Adenohipófisis/fisiología , Hormonas Hipofisarias/genética , Transactivadores/genética , Factores de Edad , Animales , Northern Blotting , Hipotiroidismo Congénito , Estradiol/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Mutantes , Factor de Transcripción PAX8 , Factores de Transcripción Paired Box , Embarazo , ARN Mensajero/análisis , Glándula Tiroides/anomalías , Hormonas Tiroideas/farmacología
14.
Cell Tissue Res ; 312(2): 175-88, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12712324

RESUMEN

Connective tissue growth factor (CTGF) is a potent fibroblast mitogen and angiogenic factor which plays an important role in wound healing, cancerogenesis and fibrotic and vascular disease. Here we explored the regulation and the cellular site of the mRNA synthesis for this growth factor in the developing mouse embryo by in situ hybridisation. Strong and persistent CTGF gene expression was limited to three types of tissue: the vascular endothelium, particularly the high-pressure part of the cardiovascular system, condensed connective tissue around bone and cartilage, and maturing layer VII neurons in the cerebral cortex. With few exceptions (late tooth bud, neuroepithelium) epithelial tissue was negative. Very transient but strong expression was observed early during formation of cartilage, in late stages during perichondral ossification, on cerebral neuroepithelium, and in several discrete stages of tooth formation, on mesenchymal precursors of odontoblasts condensing on inner dental epithelium, and later on apposing regions of ameloblast and odontoblast epithelium. Altogether, the current study suggests that CTGF performs a dual role: a continuous function in the cardiovascular system, bone and cartilage-associated mesenchyme and maturing layer VII neurons, but also a more transient function associated with the formation of cartilage, bone, tooth and cerebral nerve cells.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Inmediatas-Precoces/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Animales , Animales Recién Nacidos , Huesos/metabolismo , Sistema Cardiovascular/metabolismo , Cartílago/metabolismo , Factor de Crecimiento del Tejido Conjuntivo , Femenino , Procesamiento de Imagen Asistido por Computador , Hibridación in Situ , Ratones , Embarazo , Sondas ARN , ARN Mensajero/biosíntesis , Distribución Tisular
15.
Endocrinology ; 144(3): 777-84, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12586753

RESUMEN

Thyroid hormones are essential for a variety of developmental and metabolic processes. Congenital hypothyroidism (CHT) results in severe defects in the development of different tissues, in particular brain. As an animal model for CHT, we studied Pax8(-/-) mice, which are born without a thyroid gland. We determined the expression of iodothyronine deiodinase D1 in liver and kidney, D2 in brain and pituitary, and D3 in brain, as well as serum T(4), T(3), and rT(3) levels in Pax8(-/-) vs. control mice during the first 3 wk of life. In control mice, serum T(4) and T(3) were undetectable on the day of birth (d 0) and increased to maximum levels on d 15. In Pax8(-/-) mice, serum T(4) and T(3) remained below detection limits. Serum rT(3) was high on d 0 in both groups and rapidly decreased in Pax8(-/-), but not in control mice. Hepatic and renal D1 activities and mRNA levels were low on d 0 and increased in control mice roughly parallel to serum T(4) and T(3) levels. In Pax8(-/-) mice, tissue D1 activities and mRNA levels remained low. Cerebral D2 activities were low on d 0 and increased to maximum levels on d 15, which were approximately 10-fold higher in Pax8(-/-) than in control mice. D2 mRNA levels were higher in Pax8(-/-) than in control mice only on d 21. Cerebral D3 activities and mRNA levels were high on d 0 and showed a moderate decrease between d 3 and 15, with values slightly lower in Pax8(-/-) than in control mice. One day after the injection of 200 ng T(4) or 20 ng T(3)/g body weight, tissue deiodinase activities and mRNA levels were at least partially restored toward control levels, with the exception of cerebral D3 activity. In conclusion, these findings show dramatic age and thyroid state-dependent changes in the expression of deiodinases in central and peripheral tissues of mice during the first 3 wk of life.


Asunto(s)
Hipotiroidismo Congénito , Proteínas de Unión al ADN/deficiencia , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Yoduro Peroxidasa/genética , Proteínas Nucleares , Transactivadores/deficiencia , Envejecimiento , Animales , Encéfalo/enzimología , Proteínas de Unión al ADN/fisiología , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Trastornos del Crecimiento/genética , Hipotiroidismo/enzimología , Riñón/enzimología , Hígado/enzimología , Masculino , Ratones , Ratones Noqueados , Factor de Transcripción PAX8 , Factores de Transcripción Paired Box , Hipófisis/enzimología , ARN Mensajero/análisis , Tiroxina/sangre , Tiroxina/farmacología , Transactivadores/fisiología , Triyodotironina/sangre , Triyodotironina/farmacología , Triyodotironina Inversa/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...