Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Annu Rev Physiol ; 77: 105-29, 2015.
Article in English | MEDLINE | ID: mdl-25668018

ABSTRACT

Since its discovery nearly 30 years ago, the Hedgehog (Hh) signaling pathway has been shown to be pivotal in many developmental and pathophysiological processes in several steroidogenic tissues, including the testis, ovary, adrenal cortex, and placenta. New evidence links the evolutionarily conserved Hh pathway to the steroidogenic organs, demonstrating how Hh signaling can influence their development and homeostasis and can act in concert with steroids to mediate physiological functions. In this review, we highlight the role of the components of the Hh signaling pathway in steroidogenesis of endocrine tissues.


Subject(s)
Hedgehog Proteins/physiology , Signal Transduction/physiology , Steroids/metabolism , Adrenal Glands/metabolism , Animals , Female , Humans , Male , Ovary/metabolism , Placenta/metabolism , Pregnancy , Testis/metabolism
2.
Clin Dev Immunol ; 2013: 986859, 2013.
Article in English | MEDLINE | ID: mdl-23690829

ABSTRACT

CD40-CD40L blockade has potent immunosuppressive effects in cardiac allograft rejection but is less effective in the presence of inflammatory signals. To better understand the factors that mediate CD40-CD40L blockade-resistant rejection, we studied the effects of stimulation through glucocorticoid-induced TNFR-related protein (GITR), a costimulatory protein expressed by regulatory and effector T cells. Stimulation of CD40-/- or wild-type recipient mice treated with anti-CD40L mAb (WT+anti-CD40L) and with agonistic anti-GITR mAb resulted in cardiac allograft rejection. GITR stimulation did not induce rejection once long-term graft acceptance was established. In vitro, GITR stimulation increased proliferation of effector T cells and decreased regulatory T cell (Treg) differentiation in both treatment groups. GITR-stimulated CD40-/- recipients rejected their allografts more rapidly compared to GITR-stimulated WT+anti-CD40L recipients, and this rejection, characterized by a robust Th2 response and significant eosinophilic infiltrate, could be mediated by CD4+ T cells alone. In contrast, both CD4+ and CD8+ T cells were required to induce rejection in GITR-stimulated WT+anti-CD40L-treated recipients, and the pathology of rejection was less severe. Hence, early GITR stimulation could initiate graft rejection despite CD40 deficiency or anti-CD40L mAb treatment, though the recipient response was dependent on the mechanism of CD40-CD40L disruption.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD40 Antigens/immunology , CD40 Ligand/immunology , Glucocorticoid-Induced TNFR-Related Protein/immunology , Graft Rejection/immunology , Heart Transplantation , Animals , CD40 Antigens/antagonists & inhibitors , CD40 Antigens/genetics , CD40 Ligand/antagonists & inhibitors , CD40 Ligand/genetics , Cell Differentiation , Cell Proliferation , Glucocorticoid-Induced TNFR-Related Protein/agonists , Glucocorticoid-Induced TNFR-Related Protein/genetics , Graft Rejection/genetics , Graft Rejection/pathology , Graft Survival/immunology , Mice , Mice, Knockout , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Transplantation, Homologous
3.
Mol Endocrinol ; 27(5): 754-68, 2013 May.
Article in English | MEDLINE | ID: mdl-23518926

ABSTRACT

MicroRNAs (miRNAs) are small, endogenous, non-protein-coding RNAs that are an important means of posttranscriptional gene regulation. Deletion of Dicer, a key miRNA processing enzyme, is embryonic lethal in mice, and tissue-specific Dicer deletion results in developmental defects. Using a conditional knockout model, we generated mice lacking Dicer in the adrenal cortex. These Dicer-knockout (KO) mice exhibited perinatal mortality and failure of the adrenal cortex during late gestation between embryonic day 16.5 (E16.5) and E18.5. Further study of Dicer-KO adrenals demonstrated a significant loss of steroidogenic factor 1-expressing cortical cells that was histologically evident as early as E16.5 coincident with an increase in p21 and cleaved-caspase 3 staining in the cortex. However, peripheral cortical proliferation persisted in KO adrenals as assessed by staining of proliferating cell nuclear antigen. To further characterize the embryonic adrenals from Dicer-KO mice, we performed microarray analyses for both gene and miRNA expression on purified RNA isolated from control and KO adrenals of E15.5 and E16.5 embryos. Consistent with the absence of Dicer and the associated loss of miRNA-mediated mRNA degradation, we observed an up-regulation of a small subset of adrenal transcripts in Dicer-KO mice, most notably the transcripts coded by the genes Nr6a1 and Acvr1c. Indeed, several miRNAs, including let-7, miR-34c, and miR-21, that are predicted to target these genes for degradation, were also markedly down-regulated in Dicer-KO adrenals. Together these data suggest a role for miRNA-mediated regulation of a subset of genes that are essential for normal adrenal growth and homeostasis.


Subject(s)
Adrenal Cortex/embryology , Adrenal Cortex/metabolism , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Ribonuclease III/metabolism , Adrenal Cortex/cytology , Animals , Animals, Newborn , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Base Sequence , CD3 Complex/metabolism , Cell Cycle/genetics , Cell Death/genetics , Cell Proliferation , Conserved Sequence/genetics , DEAD-box RNA Helicases/deficiency , DNA Damage/genetics , Down-Regulation/genetics , Embryonic Development/genetics , Gene Expression Profiling , Integrases/metabolism , Mice , Mice, Knockout , MicroRNAs/genetics , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Ribonuclease III/deficiency , Software , Survival Analysis
4.
Endocr Rev ; 30(3): 241-63, 2009 May.
Article in English | MEDLINE | ID: mdl-19403887

ABSTRACT

Scientists have long hypothesized the existence of tissue-specific (somatic) stem cells and have searched for their location in different organs. The theory that adrenocortical organ homeostasis is maintained by undifferentiated stem or progenitor cells can be traced back nearly a century. Similar to other organ systems, it is widely believed that these rare cells of the adrenal cortex remain relatively undifferentiated and quiescent until needed to replenish the organ, at which time they undergo proliferation and terminal differentiation. Historical studies examining cell cycle activation by label retention assays and regenerative potential by organ transplantation experiments suggested that the adrenocortical progenitors reside in the outer periphery of the adrenal gland. Over the past decade, the Hammer laboratory, building on this hypothesis and these observations, has endeavored to understand the mechanisms of adrenocortical development and organ maintenance. In this review, we summarize the current knowledge of adrenal organogenesis. We present evidence for the existence and location of adrenocortical stem/progenitor cells and their potential contribution to adrenocortical carcinomas. Data described herein come primarily from studies conducted in the Hammer laboratory with incorporation of important related studies from other investigators. Together, the work provides a framework for the emerging somatic stem cell field as it relates to the adrenal gland.


Subject(s)
Adrenal Cortex/cytology , Stem Cells/cytology , Adrenal Cortex/embryology , Adrenal Cortex/physiology , Animals , Cell Differentiation/physiology , Clone Cells/cytology , Clone Cells/physiology , Humans , Organogenesis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...