Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Invest ; 132(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-34905516

ABSTRACT

Desmoglein 1 (Dsg1) is a cadherin restricted to stratified tissues of terrestrial vertebrates, which serve as essential physical and immune barriers. Dsg1 loss-of-function mutations in humans result in skin lesions and multiple allergies, and isolated patient keratinocytes exhibit increased proallergic cytokine expression. However, the mechanism by which genetic deficiency of Dsg1 causes chronic inflammation is unknown. To determine the systemic response to Dsg1 loss, we deleted the 3 tandem Dsg1 genes in mice. Whole transcriptome analysis of embryonic Dsg1-/- skin showed a delay in expression of adhesion/differentiation/keratinization genes at E17.5, a subset of which recovered or increased by E18.5. Comparing epidermal transcriptomes from Dsg1-deficient mice and humans revealed a shared IL-17-skewed inflammatory signature. Although the impaired intercellular adhesion observed in Dsg1-/- mice resembles that resulting from anti-Dsg1 pemphigus foliaceus antibodies, pemphigus skin lesions exhibit a weaker IL-17 signature. Consistent with the clinical importance of these findings, treatment of 2 Dsg1-deficient patients with an IL-12/IL-23 antagonist originally developed for psoriasis resulted in improvement of skin lesions. Thus, beyond impairing the physical barrier, loss of Dsg1 function through gene mutation results in a psoriatic-like inflammatory signature before birth, and treatment with a targeted therapy significantly improved skin lesions in patients.


Subject(s)
Desmoglein 1/immunology , Desmosomes/immunology , Keratinocytes/immunology , Pemphigus/immunology , Th17 Cells/immunology , Animals , Desmoglein 1/genetics , Desmosomes/genetics , Mice , Pemphigus/genetics
2.
Front Immunol ; 11: 1185, 2020.
Article in English | MEDLINE | ID: mdl-32612606

ABSTRACT

Indoleamine 2, 3-dioxygenase 1 (IDO; IDO1; INDO) is a rate-limiting enzyme that metabolizes the essential amino acid, tryptophan, into downstream kynurenines. Canonically, the metabolic depletion of tryptophan and/or the accumulation of kynurenine is the mechanism that defines how immunosuppressive IDO inhibits immune cell effector functions and/or facilitates T cell death. Non-canonically, IDO also suppresses immunity through non-enzymic effects. Since IDO targeting compounds predominantly aim to inhibit metabolic activity as evidenced across the numerous clinical trials currently evaluating safety/efficacy in patients with cancer, in addition to the recent disappointment of IDO enzyme inhibitor therapy during the phase III ECHO-301 trial, the issue of IDO non-enzyme effects have come to the forefront of mechanistic and therapeutic consideration(s). Here, we review enzyme-dependent and -independent IDO-mediated immunosuppression as it primarily relates to glioblastoma (GBM); the most common and aggressive primary brain tumor in adults. Our group's recent discovery that IDO levels increase in the brain parenchyma during advanced age and regardless of whether GBM is present, highlights an immunosuppressive synergy between aging-increased IDO activity in cells of the central nervous system that reside outside of the brain tumor but collaborate with GBM cell IDO activity inside of the tumor. Because of their potential value for the in vivo study of IDO, we also review current transgenic animal modeling systems while highlighting three new constructs recently created by our group. This work converges on the central premise that maximal immunotherapeutic efficacy in subjects with advanced cancer requires both IDO enzyme- and non-enzyme-neutralization, which is not adequately addressed by available IDO-targeting pharmacologic approaches at this time.


Subject(s)
Brain Neoplasms/immunology , Brain/enzymology , Glioblastoma/immunology , Immune Tolerance/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Animals , Brain Neoplasms/enzymology , Disease Models, Animal , Glioblastoma/enzymology , Humans , Neoplasms/enzymology , Neoplasms/immunology
4.
BMC Dev Biol ; 10: 37, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20359371

ABSTRACT

BACKGROUND: The C-terminal Eps15 homology domain-containing protein 1 (EHD1) is ubiquitously expressed and regulates the endocytic trafficking and recycling of membrane components and several transmembrane receptors. To elucidate the function of EHD1 in mammalian development, we generated Ehd1-/- mice using a Cre/loxP system. RESULTS: Both male and female Ehd1-/- mice survived at sub-Mendelian ratios. A proportion of Ehd1-/- mice were viable and showed smaller size at birth, which continued into adulthood. Ehd1-/- adult males were infertile and displayed decreased testis size, whereas Ehd1-/- females were fertile. In situ hybridization and immunohistochemistry of developing wildtype mouse testes revealed EHD1 expression in most cells of the seminiferous epithelia. Histopathology revealed abnormal spermatogenesis in the seminiferous tubules and the absence of mature spermatozoa in the epididymides of Ehd1-/- males. Seminiferous tubules showed disruption of the normal spermatogenic cycle with abnormal acrosomal development on round spermatids, clumping of acrosomes, misaligned spermatids and the absence of normal elongated spermatids in Ehd1-/- males. Light and electron microscopy analyses indicated that elongated spermatids were abnormally phagocytosed by Sertoli cells in Ehd1-/- mice. CONCLUSIONS: Contrary to a previous report, these results demonstrate an important role for EHD1 in pre- and post-natal development with a specific role in spermatogenesis.


Subject(s)
Spermatogenesis , Vesicular Transport Proteins/metabolism , Animals , Endocytosis , Female , Infertility, Male/metabolism , Male , Mice , Mice, Knockout , Testis/metabolism
5.
Genesis ; 48(5): 328-42, 2010 May.
Article in English | MEDLINE | ID: mdl-20213691

ABSTRACT

The four highly homologous members of the C-terminal EH domain-containing (EHD) protein family (EHD1-4) regulate endocytic recycling. To delineate the role of EHD4 in normal physiology and development, mice with a conditional knockout of the Ehd4 gene were generated. PCR of genomic DNA and Western blotting of organ lysates from Ehd4(-/-) mice confirmed EHD4 deletion. Ehd4(-/-) mice were viable and born at expected Mendelian ratios; however, males showed a 50% reduction in testis weight, obvious from postnatal day 31. An early (Day 10) increase in germ cell proliferation and apoptosis and a later increase in apoptosis (Day 31) were seen in the Ehd4(-/-) testis. Other defects included a progressive reduction in seminiferous tubule diameter, dysregulation of seminiferous epithelium, and head abnormalities in elongated spermatids. As a consequence, lower sperm counts and reduced fertility were observed in Ehd4(-/-) males. Interestingly, EHD protein expression was seen to be temporally regulated in the testis and EHD4 levels peaked between days 10 and 15. In the adult testis, EHD4 was highly expressed in primary spermatocytes and EHD4 deletion altered the levels of other EHD proteins in an age-dependent manner. We conclude that high levels of EHD1 in the adult Ehd4(-/-) testis functionally compensate for lack of EHD4 and prevents the development of severe fertility defects. Our results suggest a role for EHD4 in the proper development of postmitotic and postmeiotic germ cells and implicate EHD protein-mediated endocytic recycling as an important process in germ cell development and testis function.


Subject(s)
DNA-Binding Proteins/genetics , Fertility/genetics , Nuclear Proteins/genetics , Testis/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Proliferation , DNA-Binding Proteins/metabolism , Female , Fertility/physiology , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Nuclear Proteins/metabolism , Organ Size , Sperm Count , Sperm Motility/genetics , Sperm Motility/physiology , Spermatids/cytology , Spermatids/metabolism , Testis/cytology , Testis/growth & development , Time Factors
6.
Dev Dyn ; 238(10): 2614-21, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19705441

ABSTRACT

The cell nucleus must be inactivated or destroyed in order to generate feeder layers for cultured cells or to prepare recipient egg cells for nuclear transfer. Existing enucleation techniques are either cumbersome or employ toxic chemicals. Here we report a new method to enucleate cells by treatment with a psoralen and long-wave ultraviolet light. The technique is >90% efficient and causes little cytoplasmic damage to the treated cell. We have used psoralen treatment to enucleate a wide variety of cells, including eggs, sperm, HeLa cells, and fibroblasts. Colonies of human embryonic stem cells (hESCs) and human keratinocyte precursors grown on psoralen-treated feeders are indistinguishable from those grown on gamma-irradiated or mitomycin C-treated cells. Psoralen enucleation provides a rapid, simple, and non-toxic method to generate feeder cells. The technique is also useful for nuclear transfer studies in species with large eggs whose cleavage divisions are not regulated by cell-cycle checkpoints.


Subject(s)
Cell Nucleus , Furocoumarins/pharmacology , Photosensitizing Agents/pharmacology , Animals , Cell Culture Techniques , Cell Nucleus/drug effects , Cell Nucleus/radiation effects , Cells, Cultured , Female , HeLa Cells , Humans , Karyotyping , Male , Oocytes/cytology , Oocytes/drug effects , Oocytes/radiation effects , Spermatozoa/cytology , Spermatozoa/drug effects , Spermatozoa/radiation effects , Ultraviolet Rays , Xenopus laevis/embryology , Xenopus laevis/growth & development
7.
J Biol Chem ; 284(39): 26402-10, 2009 Sep 25.
Article in English | MEDLINE | ID: mdl-19640839

ABSTRACT

The Ecdysoneless (Ecd) protein is required for cell-autonomous roles in development and oogenesis in Drosophila, but the function of its evolutionarily conserved mammalian orthologs is not clear. To study the cellular function of Ecd in mammalian cells, we generated Ecd(lox/lox) mouse embryonic fibroblast cells from Ecd floxed mouse embryos. Cre-mediated deletion of Ecd in Ecd(lox/lox) mouse embryonic fibroblasts led to a proliferative block due to a delay in G(1)-S cell cycle progression; this defect was reversed by the introduction of human Ecd. Loss of Ecd led to marked down-regulation of E2F target gene expression. Furthermore, Ecd directly bound to Rb at the pocket domain and competed with E2F for binding to hypophosphorylated Rb. Our results demonstrate that mammalian Ecd plays a role in cell cycle progression via the Rb-E2F pathway.


Subject(s)
Carrier Proteins/physiology , Cell Cycle/physiology , Cell Proliferation , Fibroblasts/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Female , Fibroblasts/cytology , G1 Phase/physiology , Gene Expression , Humans , Immunoblotting , Immunoprecipitation , Male , Mice , Mice, Knockout , Mice, Transgenic , Phosphorylation , Protein Binding , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , S Phase/physiology , Time Factors
8.
J Androl ; 25(4): 506-13, 2004.
Article in English | MEDLINE | ID: mdl-15223839

ABSTRACT

Calpastatin is a naturally occurring inhibitor of calpain, a protease involved in apoptotic cell death. A testis-specific isoform of calpastatin (tCAST) has been identified that is transcribed in haploid germ cells but not in spermatocytes. To investigate the possible function(s) of tCAST, we tested the hypothesis that the ectopic expression of calpastatin in spermatocytes would suppress the death of these cells in response to an apoptosis-inducing stimulus in vivo. To this end, the 5'-flanking region of the mouse ldhc gene was linked to tCAST, and transgenic mice were generated. Immunohistochemical analysis revealed that, in contrast to control sections in which the signal for tCAST was seen in round spermatids, intense staining was visualized in pachytene spermatocytes in the transgenic animals, indicating that the strategy we used to generate the transgenic animals resulted in the ectopic expression of tCAST in spermatocytes. We then tested the effect of a short period of heating on germ cell apoptosis in the testes of wild-type and transgenic mice. Pachytene spermatocytes were the major germ cell type seen to undergo apoptosis after heat treatment. There were no differences in the number of apoptotic germ cells per seminiferous tubule between wild-type and tCAST transgenic control mice; thus, there was no apparent effect of the transgene on normal apoptosis. Heating resulted in increased numbers of TUNEL-positive germ cells in both wild-type and tCAST transgenic mice, as well as increased testicular DNA fragmentation. Heating the tCAST transgenic mouse testes resulted in significantly fewer apoptotic cells per seminiferous tubule than in wild-type mice at both 8 and 24 hours after treatment. Thus, as hypothesized, the ectopic expression of tCAST in pachytene spermatocytes suppressed germ cell apoptosis.


Subject(s)
Calcium-Binding Proteins/genetics , Spermatocytes/physiology , Testis/physiology , Animals , Apoptosis , Calcium-Binding Proteins/analysis , Glutathione Transferase/genetics , Hot Temperature , Male , Meiosis , Mice , Protein Isoforms/analysis , Protein Isoforms/genetics , Recombinant Fusion Proteins/metabolism , Testis/cytology
9.
J Androl ; 24(6): 843-52, 2003.
Article in English | MEDLINE | ID: mdl-14581510

ABSTRACT

Transcription of the mouse testis-specific lactate dehydrogenase c (mldhc) gene is limited to cells of the germinal epithelium. Cloning and analysis of the mldhc promoter revealed that a 100-bp core promoter was able to regulate testis-specific transcription in vitro and in transgenic mice. Surprisingly, expression of the reporter in transgenic testes was limited to pachytene spermatocytes, whereas native LDH-C(4) was detected in pachytene and all later germ cells. To locate additional regulatory sequence that could recapitulate the native LDH-C(4) distribution pattern, we investigated the contribution of 5' and 3' flanking sequences to the regulation of LDH-C(4) expression. We found that transcription factor YY1 binds to the mldhc promoter, that the mldhc 3' untranslated sequence does not permit a postmeiotic expression of a beta-galactosidase reporter in transgenic mice, and that native mldhc mRNA is predominately meiotic, with only a low level of postmeiotic distribution. Our results suggest that the high level of LDH-C(4) in postmeiotic cells results from mRNA and protein stability.


Subject(s)
Isoenzymes/genetics , L-Lactate Dehydrogenase/genetics , Promoter Regions, Genetic/physiology , Testis/physiology , 3' Flanking Region/physiology , 5' Flanking Region/physiology , Animals , Base Sequence/genetics , DNA-Binding Proteins/physiology , Erythroid-Specific DNA-Binding Factors , Gene Expression/physiology , Genes, Reporter , Male , Meiosis/physiology , Mice , Mice, Transgenic , Molecular Sequence Data , Protein Structure, Tertiary/physiology , Proteins/metabolism , Spermatozoa/physiology , Testis/cytology , Transcription Factors/physiology , YY1 Transcription Factor , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...