Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Immunity ; 55(4): 606-622.e6, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35358427

ABSTRACT

Lymph node (LN) stromal cells play a crucial role in LN development and in supporting adaptive immune responses. However, their origin, differentiation pathways, and transcriptional programs are still elusive. Here, we used lineage-tracing approaches and single-cell transcriptome analyses to determine origin, transcriptional profile, and composition of LN stromal and endothelial progenitors. Our results showed that all major stromal cell subsets and a large proportion of blood endothelial cells originate from embryonic Hoxb6+ progenitors of the lateral plate mesoderm (LPM), whereas lymphatic endothelial cells arise from Pax3+ progenitors of the paraxial mesoderm (PXM). Single-cell RNA sequencing revealed the existence of different Cd34+ and Cxcl13+ stromal cell subsets and showed that embryonic LNs contain proliferating progenitors possibly representing the amplifying populations for terminally differentiated cells. Taken together, our work identifies the earliest embryonic sources of LN stromal and endothelial cells and demonstrates that stromal diversity begins already during LN development.


Subject(s)
Endothelial Cells , Endothelial Cells/metabolism , Lymph Nodes , Sequence Analysis, RNA , Single-Cell Analysis , Stromal Cells , Transcription Factors/metabolism
2.
Mol Genet Genomic Med ; 8(9): e1389, 2020 09.
Article in English | MEDLINE | ID: mdl-32592340

ABSTRACT

BACKGROUND: Milroy-like disease is the diagnostic definition used for patients with phenotypes that resemble classic Milroy disease (MD) but are negative to genetic testing for FLT4. In this study, we aimed at performing a genetic characterization and biochemical analysis of VEGF-C variations found in a female proband born with congenital edema consistent with Milroy-like disease. METHODS: The proband underwent next-generation sequencing-based genetic testing for a panel of genes associated with known forms of hereditary lymphedema. Segregation analysis was performed on family members by direct sequencing. In vitro studies were performed to evaluate the role of a novel identified variant. RESULTS: Two VEGF-C variations were found in the proband, a novel p.(Ser65Arg) and a pathogenic c.148-3_148-2delCA, of paternal and maternal origin, respectively. Functional characterization of the p.(Ser65Arg) variation in vitro showed alterations in VEGF-C processing. CONCLUSIONS: Our findings reveal an interesting case in which biallelic variants in VEGF-C are found in a patient with Milroy-like lymphedema. These data expand our understanding of the etiology of congenital Milroy-like lymphedema.


Subject(s)
Alleles , Lymphedema/genetics , Vascular Endothelial Growth Factor C/genetics , Adult , Child , Female , Humans , Lymphedema/pathology , Male , Middle Aged , Mutation, Missense , Pedigree , Phenotype , Vascular Endothelial Growth Factor C/metabolism
3.
Stem Cell Reports ; 12(6): 1260-1268, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31155505

ABSTRACT

Lymph nodes (LNs) are secondary lymphoid tissues that play a critical role in filtering the lymph and promoting adaptive immune responses. Surgical resection of LNs, radiation therapy, or infections may damage lymphatic vasculature and compromise immune functions. Here, we describe the generation of functional synthetic lympho-organoids (LOs) using LN stromal progenitors and decellularized extracellular matrix-based scaffolds, two basic constituents of secondary lymphoid tissues. We show that upon transplantation at the site of resected LNs, LOs become integrated into the endogenous lymphatic vasculature and efficiently restore lymphatic drainage and perfusion. Upon immunization, LOs support the activation of antigen-specific immune responses, thus acquiring properties of native lymphoid tissues. These findings provide a proof-of-concept strategy for the development of functional lympho-organoids suitable for restoring lymphatic and immune cell functions.


Subject(s)
Cells, Immobilized , Extracellular Matrix , Lymph Nodes , Organoids , Regeneration , Tissue Scaffolds/chemistry , Animals , Cells, Immobilized/metabolism , Cells, Immobilized/transplantation , Extracellular Matrix/chemistry , Extracellular Matrix/transplantation , Lymph Nodes/metabolism , Lymph Nodes/transplantation , Mice , Mice, Transgenic , Organoids/metabolism , Organoids/transplantation
4.
Nat Commun ; 9(1): 1787, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29725010

ABSTRACT

In chronic lymphocytic leukemia (CLL), the non-hematopoietic stromal microenvironment plays a critical role in promoting tumor cell recruitment, activation, survival, and expansion. However, the nature of the stromal cells and molecular pathways involved remain largely unknown. Here, we demonstrate that leukemic B lymphocytes induce the activation of retinoid acid synthesis and signaling in the microenvironment. Inhibition of RA-signaling in stromal cells causes deregulation of genes associated with adhesion, tissue organization and chemokine secretion including the B-cell chemokine CXCL13. Notably, reducing retinoic acid precursors from the diet or inhibiting RA-signaling through retinoid-antagonist therapy prolong survival by preventing dissemination of leukemia cells into lymphoid tissues. Furthermore, mouse and human leukemia cells could be distinguished from normal B-cells by their increased expression of Rarγ2 and RXRα, respectively. These findings establish a role for retinoids in murine CLL pathogenesis, and provide new therapeutic strategies to target the microenvironment and to control disease progression.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Stromal Cells/pathology , Tretinoin/physiology , Animals , Cell Line , Chemokine CXCL13/metabolism , Coculture Techniques , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Mice, Inbred C57BL , Signal Transduction , Survival Analysis , Tretinoin/metabolism , Tumor Microenvironment
5.
J Clin Invest ; 126(7): 2452-64, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27214556

ABSTRACT

The molecular mechanisms that underlie spleen development and congenital asplenia, a condition linked to increased risk of overwhelming infections, remain largely unknown. The transcription factor TLX1 controls cell fate specification and organ expansion during spleen development, and Tlx1 deletion causes asplenia in mice. Deregulation of TLX1 expression has recently been proposed in the pathogenesis of congenital asplenia in patients carrying mutations of the gene-encoding transcription factor SF-1. Herein, we have shown that TLX1-dependent regulation of retinoic acid (RA) metabolism is critical for spleen organogenesis. In a murine model, loss of Tlx1 during formation of the splenic anlage increased RA signaling by regulating several genes involved in RA metabolism. Uncontrolled RA activity resulted in premature differentiation of mesenchymal cells and reduced vasculogenesis of the splenic primordium. Pharmacological inhibition of RA signaling in Tlx1-deficient animals partially rescued the spleen defect. Finally, spleen growth was impaired in mice lacking either cytochrome P450 26B1 (Cyp26b1), which results in excess RA, or retinol dehydrogenase 10 (Rdh10), which results in RA deficiency. Together, these findings establish TLX1 as a critical regulator of RA metabolism and provide mechanistic insights into the molecular determinants of human congenital asplenia.


Subject(s)
Homeodomain Proteins/physiology , Signal Transduction , Spleen/growth & development , Tretinoin/metabolism , Animals , Cell Differentiation , Cell Lineage , Female , Gene Deletion , Heterozygote , Homozygote , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation
6.
Blood ; 127(16): 1987-97, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-26825709

ABSTRACT

Hypoxia-inducible transcription factors (HIFs) regulate a wide array of adaptive responses to hypoxia and are often activated in solid tumors and hematologic malignancies due to intratumoral hypoxia and emerging new layers of regulation. We found that in chronic lymphocytic leukemia (CLL), HIF-1α is a novel regulator of the interaction of CLL cells with protective leukemia microenvironments and, in turn, is regulated by this interaction in a positive feedback loop that promotes leukemia survival and propagation. Through unbiased microarray analysis, we found that in CLL cells, HIF-1α regulates the expression of important chemokine receptors and cell adhesion molecules that control the interaction of leukemic cells with bone marrow and spleen microenvironments. Inactivation of HIF-1α impairs chemotaxis and cell adhesion to stroma, reduces bone marrow and spleen colonization in xenograft and allograft CLL mouse models, and prolongs survival in mice. Of interest, we found that in CLL cells, HIF-1α is transcriptionally regulated after coculture with stromal cells. Furthermore, HIF-1α messenger RNA levels vary significantly within CLL patients and correlate with the expression of HIF-1α target genes, including CXCR4, thus further emphasizing the relevance of HIF-1α expression to CLL pathogenesis.


Subject(s)
Cell Communication/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Tumor Microenvironment/genetics , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Adhesion/genetics , Chemotaxis, Leukocyte/genetics , Gene Expression Regulation, Leukemic , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spleen/metabolism , Spleen/pathology , Stromal Cells/metabolism , Stromal Cells/pathology
7.
Immunity ; 38(4): 782-91, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23601687

ABSTRACT

Secondary lymphoid organ stromal cells comprise different subsets whose origins remain unknown. Herein, we exploit a genetic lineage-tracing approach to show that splenic fibroblastic reticular cells (FRCs), follicular dendritic cells (FDCs), marginal reticular cells (MRCs), and mural cells, but not endothelial cells, originate from embryonic mesenchymal progenitors of the Nkx2-5(+)Islet1(+) lineage. This lineage include embryonic mesenchymal cells with lymphoid tissue organizer (LTo) activity capable also of supporting ectopic lymphoid-like structures and a subset of resident spleen stromal cells that proliferate and regenerate the splenic stromal microenvironment following resolution of a viral infection. These findings identify progenitor cells that generate stromal diversity in spleen development and repair and suggest the existence of multipotent stromal progenitors in the adult spleen with regenerative capacity.


Subject(s)
Dendritic Cells, Follicular/metabolism , Fibroblasts/metabolism , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Spleen/pathology , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured , Dendritic Cells, Follicular/pathology , Fibroblasts/pathology , Homeobox Protein Nkx-2.5 , Lymphocytic Choriomeningitis/physiopathology , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Regeneration , Stromal Cells/metabolism , Stromal Cells/pathology
8.
Dev Cell ; 22(5): 913-26, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22560297

ABSTRACT

The molecular determinants of spleen organogenesis and the etiology of isolated congenital asplenia (ICA), a life-threatening human condition, are unknown. We previously reported that Pbx1 deficiency causes organ growth defects including asplenia. Here, we show that mice with splenic mesenchyme-specific Pbx1 inactivation exhibit hyposplenia. Moreover, the loss of Pbx causes downregulation of Nkx2-5 and derepression of p15Ink4b in spleen mesenchymal progenitors, perturbing the cell cycle. Removal of p15Ink4b in Pbx1 spleen-specific mutants partially rescues spleen growth. By whole-exome sequencing of a multiplex kindred with ICA, we identify a heterozygous missense mutation (P236H) in NKX2-5 showing reduced transactivation in vitro. This study establishes that a Pbx/Nkx2-5/p15 regulatory module is essential for spleen development.


Subject(s)
Homeodomain Proteins/genetics , Spleen/abnormalities , Splenic Diseases/genetics , Transcription Factors/genetics , Adolescent , Amino Acid Sequence , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p15/metabolism , DNA-Binding Proteins/deficiency , Exome , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/metabolism , Humans , Infant , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation, Missense , Pedigree , Pre-B-Cell Leukemia Transcription Factor 1 , Proto-Oncogene Proteins/deficiency , Transcription Factors/deficiency , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...