Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
Development ; 151(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38958026

ABSTRACT

Thymic epithelial cells (TECs) are crucial to the ability of the thymus to generate T cells for the adaptive immune system in vertebrates. However, no in vitro system for studying TEC function exists. Overexpressing the transcription factor FOXN1 initiates transdifferentiation of fibroblasts into TEC-like cells (iTECs) that support T-cell differentiation in culture or after transplant. In this study, we have characterized iTEC programming at the cellular and molecular level in mouse to determine how it proceeds, and have identified mechanisms that can be targeted for improving this process. These data show that iTEC programming consists of discrete gene expression changes that differ early and late in the process, and that iTECs upregulate markers of both cortical and medullary TEC (cTEC and mTEC) lineages. We demonstrate that promoting proliferation enhances iTEC generation, and that Notch inhibition allows the induction of mTEC differentiation. Finally, we show that MHCII expression is the major difference between iTECs and fetal TECs. MHCII expression was improved by co-culturing iTECs with fetal double-positive T-cells. This study supports future efforts to improve iTEC generation for both research and translational uses.


Subject(s)
Cell Differentiation , Epithelial Cells , Fibroblasts , Forkhead Transcription Factors , Thymus Gland , Animals , Epithelial Cells/metabolism , Epithelial Cells/cytology , Thymus Gland/cytology , Thymus Gland/metabolism , Thymus Gland/embryology , Fibroblasts/metabolism , Fibroblasts/cytology , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Mice , Cell Proliferation , Cell Transdifferentiation , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Coculture Techniques , Receptors, Notch/metabolism
2.
Front Immunol ; 15: 1423488, 2024.
Article in English | MEDLINE | ID: mdl-39072332

ABSTRACT

T cell development in the thymus is dependent on the thymic microenvironment, in which thymic epithelial cells (TECs) are the major component. However, TECs undergo both a qualitative and quantitative loss during aging, which is believed to be the major factor responsible for age-dependent thymic atrophy. FOXN1 plays a critical role in TEC development and adult TECs maintenance. We have previously reported that intrathymic injection of a recombinant (r) protein containing murine FOXN1 and a protein transduction domain increases the number of TECs in mice, leading to enhanced thymopoiesis. However, intrathymic injection may not be an ideal choice for clinical applications. In this study, we produced a rFOXN1 fusion protein containing the N-terminal of CCR9, human FOXN1 and a protein transduction domain. When injected intravenously into 14-month-old mice, the rFOXN1 fusion protein enters the thymus and TECs, and enhances thymopoiesis, resulting in increased T cell generation in the thymus and increased number of T cells in peripheral lymphoid organ. Our results suggest that the rFOXN1 fusion protein has the potential to be used in preventing and treating T cell immunodeficiency in older adults.


Subject(s)
Forkhead Transcription Factors , Recombinant Fusion Proteins , T-Lymphocytes , Thymus Gland , Animals , Mice , Recombinant Fusion Proteins/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Thymus Gland/immunology , Thymus Gland/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Humans , Aging/immunology , Mice, Inbred C57BL , Epithelial Cells/metabolism , Epithelial Cells/immunology , Cell Differentiation
3.
J Allergy Clin Immunol Glob ; 3(3): 100267, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38800615

ABSTRACT

Background: Forkhead box protein N1 (FOXN1) transcription factor plays an essential role in the development of thymic epithelial cells, required for T-cell differentiation, maturation, and function. Biallelic pathogenic variants in FOXN1 cause severe combined immunodeficiency (SCID). More recently, heterozygous variants in FOXN1, identified by restricted gene panels, were also implicated with causing a less severe and variable immunodeficiency. Objective: We undertook longitudinal follow-up and advanced genetic investigations, including whole exome sequencing and whole genome sequencing, of newborns with a heterozygous variant in FOXN1. Methods: Five patients (3 female, 2 male) have been followed since they were first detected with low T-cell receptor excision circles during newborn screening for SCID. Patients underwent immune evaluation as well as genetic testing, including a primary immunodeficiency panel, whole exome sequencing, and whole genome sequencing in some cases. Results: Median follow-up time was 6.5 years. Initial investigations revealed low CD3+ T lymphocytes in all patients. One patient presented with extremely low lymphocyte counts and depressed phytohemagglutinin responses leading to a tentative diagnosis of SCID. Over a period of 2 years, CD3+ T-cell counts rose, although in some patients it remained borderline low. One of 5 children continues to experience recurrent upper respiratory infections and asthma episodes. The remaining are asymptomatic except for eczema in 2 of 5 cases. Lymphocyte proliferation responses to phytohemagglutinin were initially low in 3 patients but normalized by age 10 months. In 3 of 5 cases, T lymphocyte counts remain low/borderline low. Conclusion: In cases of monoallelic FOXN1 variants, using whole exome sequencing and whole genome sequencing to rule out possible other significant pathogenic variants allowed us to proceed with confidence in a conservative manner, even in extreme cases consistent with newborn screen-positive early presentation of SCID.

4.
Immunol Rev ; 322(1): 178-211, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38228406

ABSTRACT

The thymus is the primary site of T-cell development, enabling generation, and selection of a diverse repertoire of T cells that recognize non-self, whilst remaining tolerant to self- antigens. Severe congenital disorders of thymic development (athymia) can be fatal if left untreated due to infections, and thymic tissue implantation is the only cure. While newborn screening for severe combined immune deficiency has allowed improved detection at birth of congenital athymia, thymic disorders acquired later in life are still underrecognized and assessing the quality of thymic function in such conditions remains a challenge. The thymus is sensitive to injury elicited from a variety of endogenous and exogenous factors, and its self-renewal capacity decreases with age. Secondary and age-related forms of thymic dysfunction may lead to an increased risk of infections, malignancy, and autoimmunity. Promising results have been obtained in preclinical models and clinical trials upon administration of soluble factors promoting thymic regeneration, but to date no therapy is approved for clinical use. In this review we provide a background on thymus development, function, and age-related involution. We discuss disease mechanisms, diagnostic, and therapeutic approaches for primary and secondary thymic defects.


Subject(s)
Immunologic Deficiency Syndromes , T-Lymphocytes , Thymus Gland/abnormalities , Infant, Newborn , Humans , Cell Differentiation
5.
Biomedicines ; 11(10)2023 Sep 27.
Article in English | MEDLINE | ID: mdl-37893027

ABSTRACT

Although the impact of age, gender, and obesity on the skin wound healing process has been extensively studied, the data related to gender differences in aspects of skin scarring are limited. The present study performed on abdominal human intact and scar skin focused on determining gender differences in extracellular matrix (ECM) composition, dermal white adipose tissue (dWAT) accumulation, and Foxn1 expression as a part of the skin response to injury. Scar skin of men showed highly increased levels of COLLAGEN 1A1, COLLAGEN 6A3, and ELASTIN mRNA expression, the accumulation of thick collagen I-positive fibers, and the accumulation of α-SMA-positive cells in comparison to the scar skin of women. However, post-injured skin of women displayed an increase (in comparison to post-injured men's skin) in collagen III accumulation in the scar area. On the contrary, women's skin samples showed a tendency towards higher levels of adipogenic-related genes (PPARγ, FABP4, LEPTIN) than men, regardless of intact or scar skin. Intact skin of women showed six times higher levels of LEPTIN mRNA expression in comparison to men intact (p < 0.05), men post-injured (p < 0.05), or women post-injured scar (p < 0.05) skin. Higher levels of FOXN1 mRNA and protein were also detected in women than in men's skin. In conclusion, the present data confirm and extend (dWAT layer) the data related to the presence of differences between men and women in the skin, particularly in scar tissues, which may contribute to the more effective and gender-tailored improvement of skin care interventions.

6.
Front Immunol ; 14: 1261081, 2023.
Article in English | MEDLINE | ID: mdl-37868985

ABSTRACT

Thymic epithelial cells (TECs) are essential for T cell development in the thymus, yet the mechanisms governing their differentiation are not well understood. Lin28, known for its roles in embryonic development, stem cell pluripotency, and regulating cell proliferation and differentiation, is expressed in endodermal epithelial cells during embryogenesis and persists in adult epithelia, implying postnatal functions. However, the detailed expression and function of Lin28 in TECs remain unknown. In this study, we examined the expression patterns of Lin28 and its target Let-7g in fetal and postnatal TECs and discovered opposing expression patterns during postnatal thymic growth, which correlated with FOXN1 and MHCII expression. Specifically, Lin28b showed high expression in MHCIIhi TECs, whereas Let-7g was expressed in MHCIIlo TECs. Deletion of Lin28a and Lin28b specifically in TECs resulted in reduced MHCII expression and overall TEC numbers. Conversely, overexpression of Lin28a increased total TEC and thymocyte numbers by promoting the proliferation of MHCIIlo TECs. Additionally, our data strongly suggest that Lin28 and Let-7g expression is reliant on FOXN1 to some extent. These findings suggest a critical role for Lin28 in regulating the development and differentiation of TECs by modulating MHCII expression and TEC proliferation throughout thymic ontogeny and involution. Our study provides insights into the mechanisms underlying TEC differentiation and highlights the significance of Lin28 in orchestrating these processes.


Subject(s)
Epithelial Cells , Thymus Gland , Pregnancy , Female , Humans , Thymus Gland/metabolism , Epithelial Cells/metabolism , Thymocytes , Epithelium , Cell Differentiation/genetics
7.
FASEB J ; 37(10): e23171, 2023 10.
Article in English | MEDLINE | ID: mdl-37682531

ABSTRACT

Intradermal adipocytes form dermal white adipose tissue (dWAT), a unique fat depot localized in the lower layer of the dermis. However, recognition of molecular factors regulating dWAT development, homeostasis, and bioactivity is limited. Using Foxn1-/- and Foxn1+/+ mice, we demonstrated that epidermally expressed Foxn1 regulates dWAT development and defines the adipogenic capacity of dermal fibroblasts. In intact and post-wounded skin, Foxn1 contributes to the initial stimulation of dWAT adipogenesis and participates in the modulation of lipid metabolism processes. Furthermore, Foxn1 activity strengthens adipogenic processes through Bmp2 and Igf2 signaling and regulates lipid metabolism in differentiated dermal fibroblasts. The results reveal the contribution of Foxn1 to dWAT metabolism, thus identifying possible targets for modulation and regulation of dWAT in physiological and pathological processes in the skin.


Subject(s)
Adipogenesis , Adipose Tissue, White , Forkhead Transcription Factors , Gene Expression Regulation , Animals , Mice , Homeostasis , Lipid Metabolism , Forkhead Transcription Factors/metabolism
8.
Eur J Immunol ; 53(12): e2350725, 2023 12.
Article in English | MEDLINE | ID: mdl-37724048

ABSTRACT

In mammals, T-cell development depends on the activity of the Foxn1 transcription factor in the thymic epithelium; mutations in the vertebrate-specific Foxn1 gene are associated with profound T-cell lymphopenia and fatal immunodeficiency. Here, we examined the extent of T-cell development in teleosts lacking a functional foxn1 gene. In zebrafish carrying a deleterious internal deletion of foxn1, reduced but robust lymphopoietic activity is maintained in the mutant thymus. Moreover, pseudogenization or loss of foxn1 in the genomes of deep-sea anglerfishes is independent of the presence or absence of the canonical signatures of the T-cell lineage. Thus, in contrast to the situation in mammals, the teleost thymus can support foxn1-independent lymphopoiesis, most likely through the activity of the Foxn4, an ancient metazoan paralog of Foxn1. Our results imply that during the early stages of vertebrate evolution, genetic control of thymopoiesis was functionally redundant and thus robust; in mammals, the genetic network was reorganized to become uniquely dependent on the FOXN1 transcription factor.


Subject(s)
Gene Regulatory Networks , Zebrafish , Mice , Animals , Mice, Transgenic , Zebrafish/genetics , T-Lymphocytes , Thymus Gland , Transcription Factors/genetics , Forkhead Transcription Factors/genetics , Epithelial Cells , Mammals/genetics , Zebrafish Proteins/genetics
9.
Brain Behav Immun ; 113: 341-352, 2023 10.
Article in English | MEDLINE | ID: mdl-37541395

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is the most common cause of dementia in older adults and characterized by progressive loss of memory and cognitive functions that are associated with amyloid-beta (Aß) plaques and neurofibrillary tangles. Immune cells play an important role in the clearance of Aß deposits and neurofibrillary tangles. T cells are the major component of the immune system. The thymus is the primary organ for T cell generation. T cell development in the thymus depends on thymic epithelial cells (TECs). However, TECs undergo both qualitative and quantitative loss over time. We have previously reported that a recombinant (r) protein containing FOXN1 and a protein transduction domain can increase the number of TECs and subsequently increases the number of T cells in mice. In this study we determined the ability of rFOXN1 to affect cognitive performance and AD pathology in mice. METHODS: Aged 3xTg-AD and APP/PS1 AD mice were injected with rFOXN1 or control protein. Cognitive performance, AD pathology, the thymic microenvironment and immune cells were then analyzed. RESULTS: Administration of rFOXN1 into AD mice improves cognitive performance and reduces Aß plaque load and phosphorylated tau in the brain. This is related to rejuvenating the aged thymic microenvironment, which results in enhanced T cell generation in the thymus, leading to increased number of T cells, especially IFNγ-producing T cells, in the spleen and the choroid plexus (CP), enhanced expression of immune cell trafficking molecules in the CP, and increased migration of monocyte-derived macrophages into the brain. Furthermore, the production of anti-Aß antibodies in the serum and the brain, and the macrophage phagocytosis of Aß are enhanced in rFOXN1-treated AD mice. CONCLUSIONS: Our results suggest that rFOXN1 protein has the potential to provide a novel approach to treat AD patients.


Subject(s)
Alzheimer Disease , Animals , Mice , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Disease Models, Animal , Mice, Transgenic , Plaque, Amyloid/metabolism , Humans
10.
J Allergy Clin Immunol ; 152(5): 1273-1291.e15, 2023 11.
Article in English | MEDLINE | ID: mdl-37419334

ABSTRACT

BACKGROUND: Thymus hypoplasia due to stromal cell problems has been linked to mutations in several transcription factors, including Forkhead box N1 (FOXN1). FOXN1 supports T-cell development by regulating the formation and expansion of thymic epithelial cells (TECs). While autosomal recessive FOXN1 mutations result in a nude and severe combined immunodeficiency phenotype, the impact of single-allelic or compound heterozygous FOXN1 mutations is less well-defined. OBJECTIVE: With more than 400 FOXN1 mutations reported, their impact on protein function and thymopoiesis remains unclear for most variants. We developed a systematic approach to delineate the functional impact of diverse FOXN1 variants. METHODS: Selected FOXN1 variants were tested with transcriptional reporter assays and imaging studies. Thymopoiesis was assessed in mouse lines genocopying several human FOXN1 variants. Reaggregate thymus organ cultures were used to compare the thymopoietic potential of the FOXN1 variants. RESULTS: FOXN1 variants were categorized into benign, loss- or gain-of-function, and/or dominant-negatives. Dominant negative activities mapped to frameshift variants impacting the transactivation domain. A nuclear localization signal was mapped within the DNA binding domain. Thymopoiesis analyses with mouse models and reaggregate thymus organ cultures revealed distinct consequences of particular Foxn1 variants on T-cell development. CONCLUSIONS: The potential effect of a FOXN1 variant on T-cell output from the thymus may relate to its effects on transcriptional activity, nuclear localization, and/or dominant negative functions. A combination of functional assays and thymopoiesis comparisons enabled a categorization of diverse FOXN1 variants and their potential impact on T-cell output from the thymus.


Subject(s)
T-Lymphocytes , Thymus Gland , Animals , Humans , Mice , Cell Differentiation , Epithelial Cells/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Phenotype , T-Lymphocytes/metabolism
11.
Development ; 150(8)2023 04 15.
Article in English | MEDLINE | ID: mdl-36975725

ABSTRACT

The transcription factor FOXN1 is essential for fetal thymic epithelial cell (TEC) differentiation and proliferation. Postnatally, Foxn1 levels vary widely between TEC subsets, from low/undetectable in putative TEC progenitors to highest in differentiated TEC subsets. Correct Foxn1 expression is required to maintain the postnatal microenvironment; premature downregulation of Foxn1 causes a rapid involution-like phenotype, and transgenic overexpression can cause thymic hyperplasia and/or delayed involution. We investigated a K5.Foxn1 transgene that drives overexpression in mouse TECs, but causes neither hyperplasia nor delay or prevention of aging-related involution. Similarly, this transgene cannot rescue thymus size in Foxn1lacZ/lacZ mice, which undergo premature involution as a result of reduced Foxn1 levels. However, TEC differentiation and cortico-medullary organization are maintained with aging in both K5.Foxn1 and Foxn1lacZ/lacZ mice. Analysis of candidate TEC markers showed co-expression of progenitor and differentiation markers as well as increased proliferation in Plet1+ TECs associated with Foxn1 expression. These results demonstrate that the functions of FOXN1 in promoting TEC proliferation and differentiation are separable and context dependent, and suggest that modulating Foxn1 levels can regulate the balance of proliferation and differentiation in TEC progenitors.


Subject(s)
Gene Expression Regulation , Thymus Gland , Animals , Mice , Cell Differentiation/genetics , Cell Proliferation/genetics , Down-Regulation , Epithelial Cells/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Mice, Inbred C57BL
12.
Res Sq ; 2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36798162

ABSTRACT

Background: Although the thymus continues to export T cells throughout life, it undergoes a profound involution/atrophy with age, resulting in decreased numbers of T cells in the older adult, which has direct etiological linkages with many diseases. T cell development in the thymus is dependent on the thymic microenvironment, in which thymic epithelial cells (TECs) are the major component. However, TECs undergo both a qualitative and quantitative loss during aging, which is believed to be the major factor responsible for age-dependent thymic atrophy. FOXN1 plays a critical role in TEC development and adult TECs maintenance. We have previously reported that intrathymic injection of a recombinant (r) protein containing FOXN1 and a protein transduction domain increases the number of TECs in mice, leading to enhanced thymopoiesis. However, intrathymic injection may not be an ideal choice for clinical applications. In this study, we produce a rFOXN1 fusion protein containing the N-terminal of CCR9, FOXN1 and a protein transduction domain. Results: We show here that, when injected intravenously into aged mice, the rFOXN1 fusion protein migrates into the thymus and enhances thymopoiesis, resulting in increased T cell generation in the thymus and increased number of T cells in peripheral lymphoid organ. Conclusions: Our results suggest that the rFOXN1 fusion protein has the potential to be used in preventing and treating T cell immunodeficiency in the older adult.

13.
Int Immunopharmacol ; 116: 109798, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36738681

ABSTRACT

Previous studies have found that thymus is involved in the process of hypertension. However, whether thymus transplantation alleviates target organ damage in hypertensive mice remains unknown. The aim of this study was to evaluate the effects of thymus transplantation on blood pressure and target organ changes in mice with hypertension. Mice were randomly divided into normal control group (Con), hypertensive group (HTN) and thymus transplantation group (HTN + Trans). Thymus of neonatal mice was transplanted into the renal capsule of the transplantation group. After transplantation, the mouse tail noninvasive pressure was measured and heart function was evaluated weekly. Then mice were euthanized and organs or tissues were harvested at 4 weeks post-transplantation. The blood pressure of HTN + Trans group was lower than that in the HTN group. The expression of FoxN1, Aire, ATRAP, thymosin ß4 and the content of sjTREC in thymus of HTN group was decreased and the number of naïve T cells in HTN group was lower compared with other two groups. The ratio of cTEC/mTEC in HTN group was higher than that in Con group and lower than that in HTN + Trans group. Cardiac pathology showed cardiac hypertrophy and fibrosis in HTN group whereas thymus transplantation improved heart function and structure. Altogether, our findings demonstrated thymus transplantation could improve thymus function of hypertensive mice, which increased the expression of thymus transcription factor FoxN1, affected the proportion of T cell subsets, and increased thymosin ß4 thereby reducing blood pressure and reversing the progression of target organ damage.


Subject(s)
Hypertension , Animals , Mice , Blood Pressure , Kidney/pathology , Thymus Gland/pathology , Transcription Factors
14.
Mol Biol Rep ; 50(2): 1231-1239, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36441374

ABSTRACT

BACKGROUND: The Cas9 nuclease is delivered in the form of either Cas9 protein or mRNA along with CRISPR guide RNA (gRNA: dual-crRNA:tracrRNA or chimeric single-guide RNA) or in a plasmid package encoding both Cas9 and the CRISPR gRNA. METHODS AND RESULTS: We directly compared the efficiency of producing rat blastocysts with homozygous mutations of the Foxn1 locus by pronuclear injection of Cas9 in the form of protein, mRNA, or plasmid DNA. For highly efficient production of rat blastocysts with homozygous Foxn1 mutations, pronuclear injection of Cas9 protein at 60 ng/µl was likely optimal. While blastocyst harvest in the mRNA groups was higher than those in the protein and plasmid DNA groups, genotype analysis showed that 63.6%, 8.7-20.0%, and 25.0% of the analyzed blastocysts were homozygous mutants in the protein, mRNA, and plasmid DNA groups, respectively. The high efficiency of producing homozygous mutant blastocysts in the 60 ng/µl protein group may be associated with primary genome editing being initiated before the first cleavage. In most cases, homozygous mutations at the target Foxn1 locus are triggered by deletion and repair via nonhomologous end joining or microhomology-mediated end joining. Deletion downstream of the Cas9 break site was more likely than deletion in the upstream direction. CONCLUSIONS: The Cas9 nuclease in protein form, when coinjected with the CRISPR gRNA (ribonucleoprotein) into a rat zygote pronucleus, can access the target genome site and induce double-strand breaks promptly, resulting in the efficient production of homozygous mutants.


Subject(s)
CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Animals , Rats , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , Mutation/genetics , DNA , RNA, Messenger/genetics
15.
FASEB J ; 36(8): e22436, 2022 08.
Article in English | MEDLINE | ID: mdl-35792861

ABSTRACT

Skin exposed to environmental threats, including injuries and oxidative stress, develops an efficient but not fully recognized system of repair and antioxidant protection. Here, using mass spectrometry analysis (LC-MS/MS), followed by in vitro and in vivo experiments, we provided evidence that Foxn1 in keratinocytes regulates elements of the electron transport chain and participates in the thioredoxin system (Txn2, Txnrd3, and Srxn1) induction, particularly in a hypoxic environment. We first showed that Foxn1 in keratinocytes upregulates glutathione thioredoxin reductase 3 (Txnrd3) protein expression, and high levels of Txnrd3 mRNA were detected in injured skin of Foxn1+/+ mice. We also showed that Foxn1 strongly downregulated the Ccn2 protein expression, participating in epidermal reconstruction after injury. An in vitro assay revealed that Foxn1 controls keratinocyte migration, stimulating it under normoxia and suppressing it under hypoxia. Keratinocytes overexpressing Foxn1 and exposed to hypoxia displayed a reduced ability to promote angiogenesis by downregulating Vegfa expression. In conclusion, this study showed a new mechanism in which Foxn1, along with hypoxia, participates in the activation of antioxidant defense and controls the functional properties of keratinocytes.


Subject(s)
Antioxidants , Wound Healing , Animals , Antioxidants/metabolism , Chromatography, Liquid , Hypoxia/metabolism , Keratinocytes/metabolism , Mice , Tandem Mass Spectrometry , Wound Healing/physiology
16.
Front Immunol ; 13: 864777, 2022.
Article in English | MEDLINE | ID: mdl-35757725

ABSTRACT

The thymus, a primary lymphoid organ, produces the T cells of the immune system. Originating from the 3rd pharyngeal pouch during embryogenesis, this organ functions throughout life. Yet, thymopoiesis can be transiently or permanently damaged contingent on the types of systemic stresses encountered. The thymus also undergoes a functional decline during aging, resulting in a progressive reduction in naïve T cell output. This atrophy is evidenced by a deteriorating thymic microenvironment, including, but not limited, epithelial-to-mesenchymal transitions, fibrosis and adipogenesis. An exploration of cellular changes in the thymus at various stages of life, including mouse models of in-born errors of immunity and with single cell RNA sequencing, is revealing an expanding number of distinct cell types influencing thymus functions. The thymus microenvironment, established through interactions between immature and mature thymocytes with thymus epithelial cells (TEC), is well known. Less well appreciated are the contributions of neural crest cell-derived mesenchymal cells, endothelial cells, diverse hematopoietic cell populations, adipocytes, and fibroblasts in the thymic microenvironment. In the current review, we will explore the contributions of the many stromal cell types participating in the formation, expansion, and contraction of the thymus under normal and pathophysiological processes. Such information will better inform approaches for restoring thymus functionality, including thymus organoid technologies, beneficial when an individuals' own tissue is congenitally, clinically, or accidentally rendered non-functional.


Subject(s)
Endothelial Cells , Thymocytes , Adipogenesis , Animals , Epithelial Cells/metabolism , Mice , Stromal Cells , Thymocytes/metabolism , Thymus Gland
17.
Biotech Histochem ; 97(8): 584-592, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35527675

ABSTRACT

Odontogenic keratocysts (OKC) and orthokeratinized odontogenic cysts (OOC) are odontogenic cysts that share histological and immunohistochemical similarity with epidermal appendages and cutaneous cystic lesions despite exhibiting contrasting biological behavior. In epidermal appendages, BMP4 induces expression of FOXN1, which participates in terminal differentiation of keratinocytes and control of proliferation. We compared BMP4 and FOXN1 expression in OOC and OKC to investigate their role in the epithelial differentiation of these cysts. BMP4 and FOXN1 expression was assessed using immunohistochemistry in 20 primary sporadic OKC and compared to 16 OOC. BMP4 epithelial expression was detected in 81.25% OOC compared to 35% in OKC, while its expression in connective tissue was observed in 65% OKC and 75% OOC. FOXN1 was detected in 75% OOC vs. 30% OKC. The "triple positive" phenotype, i.e., BMP4 epithelial and connective tissue positivity and FOXN1 epithelial positivity, was seen in 56.25% OOC compared to 10% OKC. The greater expression of BMP4 and FOXN1 in OOC suggests greater activation of this pathway in OOC, which suggests a role in its more mature epithelium; it also resembles an epidermal phenotype.


Subject(s)
Odontogenic Cysts , Odontogenic Tumors , Humans , Bone Morphogenetic Protein 4/metabolism , Epithelium/metabolism , Immunohistochemistry , Odontogenic Cysts/pathology , Odontogenic Tumors/metabolism , Phenotype
18.
Cureus ; 14(11): e32040, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36600823

ABSTRACT

Forkheadbox N1 (FOXN1) gene mutation in humans is a rare cause of thymic hypoplasia and T cell immunodeficiency. This gene is the master transcriptional regulator of thymic epithelial cells and disruptions have been described in consequence to a variety of antepartum complications. FOXN1 mutation-mediated immune deficiency is typically associated with severe combined immunodeficiency and alopecia universalis (SCID/NUDE phenotypes) with homozygous alterations in human animal models. Less common, however, FOXN1 alterations can occur in a heterozygous form and provide a distinct phenotype of severe combined immunodeficiency (SCID) without alopecia. Here, we present one such case of a Caucasian child born with heterozygous FOXN1 mutation, first presenting with undetectable T cell levels at newborn screen. He was confirmed to have FOXN1 immunodeficiency in the heterozygous form through genetic testing. Early identification and initiation of appropriate interventions are crucial to reduce mortality from opportunistic pathogens associated with immunodeficiency. Furthermore, we need to appreciate the less common presentations of established diseases among young patients.

19.
Clin Epigenetics ; 13(1): 210, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819170

ABSTRACT

BACKGROUND: The thymic microenvironment is mainly comprised of thymic epithelial cells, the cytokines, exosomes, surface molecules, and hormones from the cells, and plays a vital role in the development, differentiation, maturation and homeostasis of T lymphocytes. However, the thymus begins to degenerate as early as the second year of life and continues through aging in human beings, leading to a decreased output of naïve T cells, the limited TCR diversity and an expansion of monoclonal memory T cells in the periphery organs. These alternations will reduce the adaptive immune response to tumors and emerging infectious diseases, such as COVID-19, also it is easier to suffer from autoimmune diseases in older people. In the context of global aging, it is important to investigate and clarify the causes and mechanisms of thymus involution. MAIN BODY: Epigenetics include histone modification, DNA methylation, non-coding RNA effects, and chromatin remodeling. In this review, we discuss how senescent thymic epithelial cells determine and control age-related thymic atrophy, how this process is altered by epigenetic modification. How the thymus adipose influences the dysfunctions of the thymic epithelial cells, and the prospects of targeting thymic epithelial cells for the treatment of thymus atrophy. CONCLUSION: Epigenetic modifications are emerging as key regulators in governing the development and senescence of thymic epithelial cells. It is beneficial to re-establish effective thymopoiesis, identify the potential therapeutic strategy and rejuvenate the immune function in the elderly.


Subject(s)
Aging/physiology , Epigenesis, Genetic/physiology , Epithelial Cells/pathology , Thymus Gland/pathology , Atrophy , Humans
20.
Front Immunol ; 12: 652665, 2021.
Article in English | MEDLINE | ID: mdl-33859647

ABSTRACT

T cell development is effectively supported in fetal thymus organ cultures (FTOCs), which places thymus lobes atop an air-liquid interface (ALI) culture system. The direct exposure to air is critical for its success, as fetal thymus lobes placed in low oxygen submersion (LOS)-FTOCs fail to support thymocyte development. However, submersion cultures performed in the presence of high concentration of ambient oxygen (60~80%) allow for normal thymocyte development, but the underlying mechanism for this rescue has remained elusive. Here, we show that FOXN1 expression in thymic epithelial cells (TECs) from LOS-FTOCs was greatly reduced compared to conventional ALI-FTOCs. Consequently, the expression of important FOXN1 target genes, including Dll4 and Ccl25, in TECs was extinguished. The loss of DLL4 and CCL25 interrupted thymocyte differentiation and led to CD4+CD8+ cells exiting the lobes, respectively. High oxygen submersion (HOS)-FTOCs restored the expression of FOXN1 and its target genes, as well as maintained high levels of MHCII expression in TECs. In addition, HOS-FTOCs promoted the self-renewal of CD4-CD8-CD44-CD25+ cells, allowing for the continuous generation of later stage thymocytes. Forced FOXN1 expression in TECs rescued thymocyte developmental progression, but not cellularity, in LOS-FTOCs. Given that oxidative stress has been reported to accelerate the onset of age-associated thymic involution, we postulate that regulation of FOXN1 by oxygen and antioxidants may underpin this biological process.


Subject(s)
Forkhead Transcription Factors/metabolism , Lymphopoiesis , Organ Culture Techniques , Oxygen/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Animals , Biomarkers , Cell Differentiation , Immunophenotyping , Mice , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL