Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Semin Intervent Radiol ; 40(3): 269-273, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37484444

ABSTRACT

Transjugular intrahepatic portosystemic shunt (TIPS) is an important interventional option for the treatment of complications related to cirrhosis and portal hypertension. Emergent TIPS placement can be a life-saving measure in patients with uncontrolled variceal hemorrhage. After TIPS placement, patients may benefit from additional interventions for clinical optimization including stent dilation, stent extension, and embolization of varices. Here, we describe a case of emergent TIPS placement and revision which resulted in TIPS stent migration requiring stent removal and replacement. We discuss our technique and review previously reported methods for the management of migrated TIPS stents.

3.
Semin Intervent Radiol ; 37(4): 356-364, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33041481

ABSTRACT

Endovascular aneurysmal repair (EVAR) has become a prominent modality for the treatment of abdominal aortic aneurysm. Surveillance imaging is important for the detection of device-related complications, which include endoleak, structural abnormalities, and infection. Currently used modalities include ultrasound, X-ray, computed tomography, magnetic resonance imaging, and angiography. Understanding the advantages and drawbacks of each modality, as well available guidelines, can guide selection of the appropriate technique for individual patients. We review complications following EVAR and advances in surveillance imaging modalities.

4.
Sci Rep ; 9(1): 11434, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31391476

ABSTRACT

The highly conserved SNARE protein SEC22B mediates diverse and critical functions, including phagocytosis, cell growth, autophagy, and protein secretion. However, these characterizations have thus far been limited to in vitro work. Here, we expand our understanding of the role Sec22b plays in vivo. We utilized Cre-Lox mice to delete Sec22b in three tissue compartments. With a germline deletion of Sec22b, we observed embryonic death at E8.5. Hematopoietic/endothelial cell deletion of Sec22b also resulted in in utero death. Notably, mice with Sec22b deletion in CD11c-expressing cells of the hematopoietic system survive to adulthood. These data demonstrate Sec22b contributes to early embryogenesis through activity both in hematopoietic/endothelial tissues as well as in other tissues yet to be defined.


Subject(s)
Embryonic Development , Endothelial Cells/metabolism , Hematopoietic System/embryology , R-SNARE Proteins/metabolism , Animals , Embryo, Mammalian , Female , Male , Mice , Mice, Knockout , R-SNARE Proteins/genetics
5.
J Clin Invest ; 129(5): 2029-2042, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30958798

ABSTRACT

DCs undergo metabolic reprogramming from a predominantly oxidative phosphorylation (OXPHOS) to glycolysis to mount an immunogenic response. The mechanism underpinning the metabolic reprogramming remains elusive. We demonstrate that miRNA-142 (miR-142) is pivotal for this shift in metabolism, which regulates the tolerogenic and immunogenic responses of DCs. In the absence of miR-142, DCs fail to switch from OXPHOS and show reduced production of proinflammatory cytokines and the ability to activate T cells in vitro and in in vivo models of sepsis and alloimmunity. Mechanistic studies demonstrate that miR-142 regulates fatty acid (FA) oxidation, which causes the failure to switch to glycolysis. Loss- and gain-of-function experiments identified carnitine palmitoyltransferase -1a (CPT1a), a key regulator of the FA pathway, as a direct target of miR-142 that is pivotal for the metabolic switch. Thus, our findings show that miR-142 is central to the metabolic reprogramming that specifically favors glycolysis and immunogenic response by DCs.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/metabolism , MicroRNAs/metabolism , Oxidative Phosphorylation , Animals , Bone Marrow Transplantation , Carnitine O-Palmitoyltransferase/metabolism , Endotoxins/metabolism , Fatty Acids/metabolism , Flow Cytometry , Glucose/metabolism , Glycolysis , Inflammation , Lipopolysaccharides/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Spleen/metabolism , T-Lymphocytes/cytology , Toll-Like Receptor 4/metabolism
6.
J Immunol ; 200(12): 4012-4023, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29703862

ABSTRACT

Maintenance of the regulatory T (Treg) cell pool is essential for peripheral tolerance and prevention of autoimmunity. Integrins, heterodimeric transmembrane proteins consisting of α and ß subunits that mediate cell-to-cell and cell-to-extracellular matrix interactions, play an important role in facilitating Treg cell contact-mediated suppression. In this article, we show that integrin activation plays an essential, previously unappreciated role in maintaining murine Treg cell function. Treg cell-specific loss of talin, a ß integrin-binding protein, or expression of talin(L325R), a mutant that selectively abrogates integrin activation, resulted in lethal systemic autoimmunity. This dysfunction could be attributed, in part, to a global dysregulation of the Treg cell transcriptome. Activation of integrin α4ß1 led to increased suppressive capacity of the Treg cell pool, suggesting that modulating integrin activation on Treg cells may be a useful therapeutic strategy for autoimmune and inflammatory disorders. Taken together, these results reveal a critical role for integrin-mediated signals in controlling peripheral tolerance by virtue of maintaining Treg cell function.


Subject(s)
Integrins/immunology , Peripheral Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/immunology , Inflammation/immunology , Mice , Talin/immunology , Transcriptome/immunology
7.
J Clin Invest ; 127(10): 3609-3623, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28846070

ABSTRACT

During an immune response, CD8+ T lymphocytes can undergo asymmetric division, giving rise to daughter cells that exhibit distinct tendencies to adopt terminal effector and memory cell fates. Here we show that "pre-effector" and "pre-memory" cells resulting from the first CD8+ T cell division in vivo exhibited low and high rates of endogenous proteasome activity, respectively. Pharmacologic reduction of proteasome activity in CD8+ T cells early during differentiation resulted in acquisition of terminal effector cell characteristics, whereas enhancement of proteasome activity conferred attributes of memory lymphocytes. Transcriptomic and proteomic analyses revealed that modulating proteasome activity in CD8+ T cells affected cellular metabolism. These metabolic changes were mediated, in part, through differential expression of Myc, a transcription factor that controls glycolysis and metabolic reprogramming. Taken together, these results demonstrate that proteasome activity is an important regulator of CD8+ T cell fate and raise the possibility that increasing proteasome activity may be a useful therapeutic strategy to enhance the generation of memory lymphocytes.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Division/immunology , Glycolysis/immunology , Immunologic Memory , Proteasome Endopeptidase Complex/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Mice , Mice, Mutant Strains , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-myc/immunology , Proto-Oncogene Proteins c-myc/metabolism
8.
Cell Rep ; 19(13): 2645-2656, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28658614

ABSTRACT

Cross-presentation initiates immune responses against tumors and viral infections by presenting extracellular antigen on MHC I to activate CD8+ T cell-mediated cytotoxicity. In vitro studies in dendritic cells (DCs) established SNARE protein SEC22B as a specific regulator of cross-presentation. However, the in vivo contribution of SEC22B to cross-presentation has not been tested. To address this, we generated DC-specific Sec22b knockout (CD11c-Cre Sec22bfl/fl) mice. Contrary to the paradigm, SEC22B-deficient DCs efficiently cross-present both in vivo and in vitro. Although in vitro small hairpin RNA (shRNA)-mediated Sec22b silencing in bone-marrow-derived dendritic cells (BMDCs) reduced cross-presentation, treatment of SEC22B-deficient BMDCs with the same shRNA produced a similar defect, suggesting the Sec22b shRNA modulates cross-presentation through off-target effects. RNA sequencing of Sec22b shRNA-treated SEC22B-deficient BMDCs demonstrated several changes in the transcriptome. Our data demonstrate that contrary to the accepted model, SEC22B is not necessary for cross-presentation, cautioning against extrapolating phenotypes from knockdown studies alone.


Subject(s)
Antigen Presentation/immunology , R-SNARE Proteins/immunology , Animals , Cross-Priming/immunology , Mice , Mice, Transgenic
9.
J Immunol ; 198(12): 4639-4651, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28515282

ABSTRACT

Talin, a cytoskeletal protein essential in mediating integrin activation, has been previously shown to be involved in the regulation of T cell proliferation and function. In this study, we describe a role for talin in maintaining the homeostasis and survival of the regulatory T (Treg) cell pool. T cell-specific deletion of talin in Tln1fl/flCd4Cre mice resulted in spontaneous lymphocyte activation, primarily due to numerical and functional deficiencies of Treg cells in the periphery. Peripheral talin-deficient Treg cells were unable to maintain high expression of IL-2Rα, resulting in impaired IL-2 signaling and ultimately leading to increased apoptosis through downregulation of prosurvival proteins Bcl-2 and Mcl-1. The requirement for talin in maintaining high IL-2Rα expression by Treg cells was due, in part, to integrin LFA-1-mediated interactions between Treg cells and dendritic cells. Collectively, our data suggest a critical role for talin in Treg cell-mediated maintenance of immune homeostasis.


Subject(s)
Homeostasis , Lymphocyte Activation , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Talin/metabolism , Animals , Apoptosis , Dendritic Cells/immunology , Genes, bcl-2 , Interleukin-2/immunology , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Myeloid Cell Leukemia Sequence 1 Protein/genetics , T-Lymphocytes, Regulatory/physiology , Talin/deficiency , Talin/immunology
10.
Nat Immunol ; 18(4): 422-432, 2017 04.
Article in English | MEDLINE | ID: mdl-28218746

ABSTRACT

During microbial infection, responding CD8+ T lymphocytes differentiate into heterogeneous subsets that together provide immediate and durable protection. To elucidate the dynamic transcriptional changes that underlie this process, we applied a single-cell RNA-sequencing approach and analyzed individual CD8+ T lymphocytes sequentially throughout the course of a viral infection in vivo. Our analyses revealed a striking transcriptional divergence among cells that had undergone their first division and identified previously unknown molecular determinants that controlled the fate specification of CD8+ T lymphocytes. Our findings suggest a model for the differentiation of terminal effector cells initiated by an early burst of transcriptional activity and subsequently refined by epigenetic silencing of transcripts associated with memory lymphocytes, which highlights the power and necessity of single-cell approaches.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Epigenesis, Genetic , Transcription, Genetic , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cluster Analysis , Computational Biology/methods , Gene Expression Profiling , Gene Silencing , Genetic Heterogeneity , Histones/metabolism , Immunologic Memory/genetics , Immunologic Memory/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Sequence Analysis, RNA , Single-Cell Analysis , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcriptome
11.
Oncotarget ; 7(16): 21527-41, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-26930717

ABSTRACT

The epithelial-mesenchymal transition (EMT) endows carcinoma cells with phenotypic plasticity that can facilitate the formation of cancer stem cells (CSCs) and contribute to the metastatic cascade. While there is substantial support for the role of EMT in driving cancer cell dissemination, less is known about the intracellular molecular mechanisms that govern formation of CSCs via EMT. Here we show that ß2 and ß5 proteasome subunit activity is downregulated during EMT in immortalized human mammary epithelial cells. Moreover, selective proteasome inhibition enabled mammary epithelial cells to acquire certain morphologic and functional characteristics reminiscent of cancer stem cells, including CD44 expression, self-renewal, and tumor formation. Transcriptomic analyses suggested that proteasome-inhibited cells share gene expression signatures with cells that have undergone EMT, in part, through modulation of the TGF-ß signaling pathway. These findings suggest that selective downregulation of proteasome activity in mammary epithelial cells can initiate the EMT program and acquisition of a cancer stem cell-like phenotype. As proteasome inhibitors become increasingly used in cancer treatment, our findings highlight a potential risk of these therapeutic strategies and suggest a possible mechanism by which carcinoma cells may escape from proteasome inhibitor-based therapy.


Subject(s)
Down-Regulation , Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Biocatalysis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line , Cell Line, Transformed , Female , Gene Expression Profiling/methods , Humans , Kaplan-Meier Estimate , Mice, Nude , Proteasome Inhibitors/pharmacology , Transplantation, Heterologous
12.
Sci Rep ; 6: 19182, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26765121

ABSTRACT

Naïve CD8(+) T lymphocytes responding to microbial pathogens give rise to effector T cells that provide acute defense and memory T cells that provide long-lived immunity. Upon activation, CD8(+) T lymphocytes can undergo asymmetric division, unequally distributing factors to the nascent daughter cells that influence their eventual fate towards the effector or memory lineages. Individual loss of either atypical protein kinase C (aPKC) isoform, PKCζ or PKCλ/ι, partially impairs asymmetric divisions and increases CD8(+) T lymphocyte differentiation toward a long-lived effector fate at the expense of memory T cell formation. Here, we show that deletion of both aPKC isoforms resulted in a deficit in asymmetric divisions, increasing the proportion of daughter cells that inherit high amounts of effector fate-associated molecules, IL-2Rα, T-bet, IFNγR, and interferon regulatory factor 4 (IRF4). However, unlike CD8(+) T cells deficient in only one aPKC isoform, complete loss of aPKC unexpectedly increased CD8(+) T cell differentiation toward a short-lived, terminal effector fate, as evidenced by increased rates of apoptosis and decreased expression of Eomes and Bcl2 early during the immune response. Together, these results provide evidence for an important role for asymmetric division in CD8(+) T lymphocyte fate specification by regulating the balance between effector and memory precursors at the initiation of the adaptive immune response.


Subject(s)
Asymmetric Cell Division , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/physiology , Cell Differentiation , Protein Kinase C/metabolism , Adaptive Immunity , Animals , Gene Knockout Techniques , Immunologic Memory , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Protein Kinase C/deficiency , Protein Kinase C/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/physiology
13.
Plast Reconstr Surg ; 135(6): 990e-999e, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26017615

ABSTRACT

BACKGROUND: Little is known about the role of osteoclasts in cranial suture fusion. Osteoclasts are predominantly regulated by receptor activator of nuclear factor kappa B and receptor activator of nuclear factor kappa B ligand, both of which lead to osteoclast differentiation, activation, and survival; and osteoprotegerin, a soluble inhibitor of receptor activator of nuclear factor kappa B. The authors' work examines the role of osteoprotegerin in this process using knockout technology. METHODS: Wild-type, osteoprotegerin-heterozygous, and osteoprotegerin-knockout mice were imaged by serial micro-computed tomography at 3, 5, 7, 9, and 16 weeks. Suture density measurements and craniometric analysis were performed at these same time points. Posterofrontal sutures were harvested from mice after the week-16 time point and analyzed by means of histochemistry. RESULTS: Micro-computed tomographic analysis of the posterofrontal suture revealed reduced suture fusion in osteoprotegerin-knockout mice compared with wild-type and heterozygous littermates. Osteoprotegerin deficiency resulted in a statistically significant decrease in suture bone density in knockout mice. There was no reduction in the density of non-suture-containing calvarial bone between wild-type and osteoprotegerin-knockout mice. Histochemistry of suture sections supported these micro-computed tomographic findings. Finally, osteoprotegerin-knockout mice had reduced anteroposterior skull distance at all time points and an increased interorbital distance at the week-16 time point. CONCLUSION: The authors' data suggest that perturbations in the expression of osteoprotegerin and subsequent changes in osteoclastogenesis lead to alterations in murine cranial and posterofrontal suture morphology.


Subject(s)
Craniosynostoses/metabolism , Craniosynostoses/pathology , Osteoclasts/metabolism , Osteoprotegerin/deficiency , X-Ray Microtomography , Animals , Animals, Newborn , Cranial Sutures/diagnostic imaging , Cranial Sutures/metabolism , Craniosynostoses/diagnostic imaging , Frontal Bone/diagnostic imaging , Frontal Bone/physiopathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Osteoprotegerin/metabolism , Random Allocation , Sensitivity and Specificity , Tissue and Organ Harvesting
14.
J Immunol ; 194(6): 2551-60, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25667416

ABSTRACT

The link between autoimmune diseases and primary immunodeficiency syndromes has been increasingly appreciated. Immunologic evaluation of a young man with autoimmune enterocolopathy and unexplained infections revealed evidence of immunodeficiency, including IgG subclass deficiency, impaired Ag-induced lymphocyte proliferation, reduced cytokine production by CD8(+) T lymphocytes, and decreased numbers of NK cells. Genetic evaluation identified haploinsufficiency of NFAT5, a transcription factor regulating immune cell function and cellular adaptation to hyperosmotic stress, as a possible cause of this syndrome. Inhibition or deletion of NFAT5 in normal human and murine cells recapitulated several of the immune deficits identified in the patient. These results provide evidence of a primary immunodeficiency disorder associated with organ-specific autoimmunity linked to NFAT5 deficiency.


Subject(s)
Autoimmune Diseases/immunology , Gastrointestinal Diseases/immunology , Haploinsufficiency/immunology , Immunologic Deficiency Syndromes/immunology , Transcription Factors/immunology , Animals , Autoimmune Diseases/diagnosis , Autoimmune Diseases/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , DNA Mutational Analysis , Gastrointestinal Diseases/diagnosis , Gastrointestinal Diseases/genetics , Gene Expression/immunology , Haploinsufficiency/genetics , Humans , Immunoblotting , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Jurkat Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Mice, 129 Strain , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Young Adult
15.
J Immunol ; 194(5): 2249-59, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25617472

ABSTRACT

During an immune response against a microbial pathogen, activated naive T lymphocytes give rise to effector cells that provide acute host defense and memory cells that provide long-lived immunity. It has been shown that T lymphocytes can undergo asymmetric division, enabling the daughter cells to inherit unequal amounts of fate-determining proteins and thereby acquire distinct fates from their inception. In this study, we show that the absence of the atypical protein kinase C (PKC) isoforms, PKCζ and PKCλ/ι, disrupts asymmetric CD8(+) T lymphocyte division. These alterations were associated with aberrant acquisition of a pre-effector transcriptional program, detected by single-cell gene expression analyses, in lymphocytes that had undergone their first division in vivo and enhanced differentiation toward effector fates at the expense of memory fates. Together, these results demonstrate a role for atypical PKC in regulating asymmetric division and the specification of divergent CD8(+) T lymphocyte fates early during an immune response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Division/immunology , Immunity, Innate , Isoenzymes/immunology , Listeriosis/immunology , Protein Kinase C/immunology , Animals , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation , Dendritic Cells/immunology , Dendritic Cells/microbiology , Dendritic Cells/pathology , Gene Expression Regulation , Immunologic Memory , Isoenzymes/genetics , Isoenzymes/metabolism , Listeria monocytogenes/immunology , Listeriosis/enzymology , Listeriosis/microbiology , Listeriosis/pathology , Mice , Mice, Knockout , Protein Kinase C/genetics , Protein Kinase C/metabolism , Signal Transduction , Single-Cell Analysis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , T-Lymphocyte Subsets/pathology
16.
Semin Intervent Radiol ; 32(1): 54-6, 2015 Mar.
Article in English | MEDLINE | ID: mdl-27053829

ABSTRACT

Percutaneous transhepatic biliary drainage (PTBD) is a well-established and safe technique for the management of biliary obstructions and leaks. While approach is variable based on operator preference, patient anatomy, and indications; PTBD is commonly performed via a right-sided intercostal route. With a right-sided approach, pleural complications may be encountered. The authors describe a case of a right PTBD complicated by a leak into the pleural space, with the subsequent development of bilothorax.

17.
Nat Immunol ; 15(4): 365-372, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24584088

ABSTRACT

T lymphocytes responding to microbial infection give rise to effector cells that mediate acute host defense and memory cells that provide long-lived immunity, but the fundamental question of when and how these cells arise remains unresolved. Here we combined single-cell gene-expression analyses with 'machine-learning' approaches to trace the transcriptional 'roadmap' of individual CD8(+) T lymphocytes throughout the course of an immune response in vivo. Gene-expression signatures predictive of eventual fates could be discerned as early as the first T lymphocyte division and may have been influenced by asymmetric partitioning of the receptor for interleukin 2 (IL-2Rα) during mitosis. Our findings emphasize the importance of single-cell analyses in understanding fate determination and provide new insights into the specification of divergent lymphocyte fates early during an immune response to microbial infection.


Subject(s)
Adaptive Immunity , CD8-Positive T-Lymphocytes/immunology , Gene Expression Profiling/methods , Infections/immunology , Infections/microbiology , Receptors, Interleukin-2/metabolism , Single-Cell Analysis/methods , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/genetics , Cell Lineage/genetics , Computer Simulation , Listeria monocytogenes/genetics , Listeria monocytogenes/immunology , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitosis/genetics , Mitosis/immunology , Ovalbumin/genetics , Ovalbumin/immunology , Receptors, Interleukin-2/genetics , T-Lymphocyte Subsets/microbiology , T-Lymphocyte Subsets/virology , Transcriptional Activation/immunology
18.
PLoS One ; 8(9): e73086, 2013.
Article in English | MEDLINE | ID: mdl-24019898

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent progenitors that can undergo osteogenic differentiation under proper stimuli. We demonstrated that BMP9 is one of the most osteogenic BMPs. However, the molecular mechanism underlying BMP9-initiated osteogenic signaling in MSCs remains unclear. Through gene expression profiling analysis we identified several candidate mediators of BMP9 osteogenic signaling. Here, we focus on one such signaling mediator and investigate the functional role of cysteine-rich with EGF-like domains 2 (Creld2) in BMP9-initiated osteogenic signaling. Creld2 was originally identified as an ER stress-inducible factor localized in the ER-Golgi apparatus. Our genomewide expression profiling analysis indicates that Creld2 is among the top up-regulated genes in BMP9-stimulated MSCs. We confirm that Creld2 is up-regulated by BMP9 in MSCs. ChIP analysis indicates that Smad1/5/8 directly binds to the Creld2 promoter in a BMP9-dependent fashion. Exogenous expression of Creld2 in MSCs potentiates BMP9-induced early and late osteogenic markers, and matrix mineralization. Conversely, silencing Creld2 expression inhibits BMP9-induced osteogenic differentiation. In vivo stem cell implantation assay reveals that exogenous Creld2 promotes BMP9-induced ectopic bone formation and matrix mineralization, whereas silencing Creld2 expression diminishes BMP9-induced bone formation and matrix mineralization. We further show that Creld2 is localized in ER and the ER stress inducers potentiate BMP9-induced osteogenic differentiation. Our results strongly suggest that Creld2 may be directly regulated by BMP9 and ER stress response may play an important role in regulating osteogenic differentiation.


Subject(s)
Cell Adhesion Molecules/physiology , Cell Differentiation/physiology , Endoplasmic Reticulum/metabolism , Extracellular Matrix Proteins/physiology , Growth Differentiation Factor 2/physiology , Mesenchymal Stem Cells/cytology , Animals , Cell Adhesion Molecules/genetics , Cell Line , Extracellular Matrix Proteins/genetics , Gene Silencing , Humans , Mice , Reverse Transcriptase Polymerase Chain Reaction
19.
J Cell Mol Med ; 17(9): 1160-72, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23844832

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent progenitors, which give rise to several lineages, including bone, cartilage and fat. Epidermal growth factor (EGF) stimulates cell growth, proliferation and differentiation. EGF acts by binding with high affinity to epidermal growth factor receptor (EGFR) on the cell surface and stimulating the intrinsic protein tyrosine kinase activity of its receptor, which initiates a signal transduction cascade causing a variety of biochemical changes within the cell and regulating cell proliferation and differentiation. We have identified BMP9 as one of the most osteogenic BMPs in MSCs. In this study, we investigate if EGF signalling cross-talks with BMP9 and regulates BMP9-induced osteogenic differentiation. We find that EGF potentiates BMP9-induced early and late osteogenic markers of MSCs in vitro, which can be effectively blunted by EGFR inhibitors Gefitinib and Erlotinib or receptor tyrosine kinase inhibitors AG-1478 and AG-494 in a dose- and time-dependent manner. Furthermore, EGF significantly augments BMP9-induced bone formation in the cultured mouse foetal limb explants. In vivo stem cell implantation experiment reveals that exogenous expression of EGF in MSCs can effectively potentiate BMP9-induced ectopic bone formation, yielding larger and more mature bone masses. Interestingly, we find that, while EGF can induce BMP9 expression in MSCs, EGFR expression is directly up-regulated by BMP9 through Smad1/5/8 signalling pathway. Thus, the cross-talk between EGF and BMP9 signalling pathways in MSCs may underline their important roles in regulating osteogenic differentiation. Harnessing the synergy between BMP9 and EGF should be beneficial for enhancing osteogenesis in regenerative medicine.


Subject(s)
Cell Differentiation , Epidermal Growth Factor/metabolism , Growth Differentiation Factor 2/metabolism , Mesenchymal Stem Cells/cytology , Osteogenesis , Receptor Cross-Talk , Signal Transduction , Alkaline Phosphatase/metabolism , Animals , Biomarkers/metabolism , Bone Matrix/drug effects , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Choristoma/pathology , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Extremities/embryology , Fetus/drug effects , Fetus/metabolism , Growth Differentiation Factor 2/pharmacology , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Mice , Osteogenesis/drug effects , Receptor Cross-Talk/drug effects , Signal Transduction/drug effects
20.
Cancer Lett ; 336(1): 222-30, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23665505

ABSTRACT

Osteosarcoma (OS) is the most common primary malignancy of bone. We investigated the roles of insulin-like growth factor binding protein 5 (IGFBP5) domains in modulating OS tumorigenicity and metastasis. The N-terminal (to a lesser extent the C-terminal) domain inhibited cell proliferation and induced apoptosis while the C-terminal domain inhibited cell migration and invasion. The Linker domain had no independent effects. In vivo, the N-terminal domain decreased tumor growth without affecting pulmonary metastases while the C-terminal domain inhibited tumor growth and metastases. In summary, the N- and C-terminal domains modulated OS tumorigenic phenotypes while the C-terminal domain inhibited OS metastatic phenotypes.


Subject(s)
Bone Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor Binding Protein 5/metabolism , Osteosarcoma/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , HEK293 Cells , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Phenotype , Protein Interaction Domains and Motifs , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...