Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michele Rhodes is active.

Publication


Featured researches published by Michele Rhodes.


Immunity | 2000

On the Dynamics of TCR:CD3 Complex Cell Surface Expression and Downmodulation

Haiyan Liu; Michele Rhodes; David L. Wiest; Dario A. A. Vignali

TCR downmodulation following ligation by MHC:peptide complexes is considered to be a pivotal event in T cell activation. Here, we analyzed the dynamics of TCR:CD3 cell surface expression on resting and antigen-activated T cells. We show that the TCR:CD3 complex is very stable and is rapidly internalized and recycled in resting T cells. Surprisingly, the internalization rate is not increased following TCR ligation by MHC:peptide complexes, despite significant TCR downmodulation, suggesting that constitutive internalization rather than ligation-induced downmodulation serves as the force that drives serial ligation. Furthermore, TCR downmodulation is mediated by the intracellular retention of ligated complexes and degradation by lysosomes and proteasomes. Thus, our data demonstrate that ligation induces TCR downmodulation by preventing recycling rather than inducing internalization.


Immunity | 2009

Marked Induction of the Helix-Loop-Helix Protein Id3 Promotes the γδ T Cell Fate and Renders Their Functional Maturation Notch Independent

Jens Peter Holst Lauritsen; Gladys W. Wong; Sang-Yun Lee; Juliette M. Lefebvre; Maria Ciofani; Michele Rhodes; Dietmar J. Kappes; Juan Carlos Zúñiga-Pflücker; David L. Wiest

alphabeta and gammadelta T cells arise from a common thymocyte progenitor during development in the thymus. Emerging evidence suggests that the pre-T cell receptor (pre-TCR) and gammadelta T cell receptor (gammadeltaTCR) play instructional roles in specifying the alphabeta and gammadelta T-lineage fates, respectively. Nevertheless, the signaling pathways differentially engaged to specify fate and promote the development of these lineages remain poorly understood. Here, we show that differential activation of the extracellular signal-related kinase (ERK)-early growth response gene (Egr)-inhibitor of DNA binding 3 (Id3) pathway plays a defining role in this process. In particular, Id3 expression served to regulate adoption of the gammadelta fate. Moreover, Id3 was both necessary and sufficient to enable gammadelta-lineage cells to differentiate independently of Notch signaling and become competent IFNgamma-producing effectors. Taken together, these findings identify Id3 as a central player that controls both adoption of the gammadelta fate and its maturation in the thymus.


Blood | 2012

Inactivation of ribosomal protein L22 promotes transformation by induction of the stemness factor, Lin28B

Shuyun Rao; Sang-Yun Lee; Alejandro Gutierrez; Jacqueline Perrigoue; Roshan J. Thapa; Zhigang Tu; John R. Jeffers; Michele Rhodes; Stephen J. Anderson; Tamas Oravecz; Stephen P. Hunger; Roman A. Timakhov; Rugang Zhang; Siddharth Balachandran; Gerard P. Zambetti; Joseph R. Testa; A. Thomas Look; David L. Wiest

Ribosomal protein (RP) mutations in diseases such as 5q- syndrome both disrupt hematopoiesis and increase the risk of developing hematologic malignancy. However, the mechanism by which RP mutations increase cancer risk has remained an important unanswered question. We show here that monoallelic, germline inactivation of the ribosomal protein L22 (Rpl22) predisposes T-lineage progenitors to transformation. Indeed, RPL22 was found to be inactivated in ∼ 10% of human T-acute lymphoblastic leukemias. Moreover, monoallelic loss of Rpl22 accelerates development of thymic lymphoma in both a mouse model of T-cell malignancy and in acute transformation assays in vitro. We show that Rpl22 inactivation enhances transformation potential through induction of the stemness factor, Lin28B. Our finding that Rpl22 inactivation promotes transformation by inducing expression of Lin28B provides the first insight into the mechanistic basis by which mutations in Rpl22, and perhaps some other RP genes, increases cancer risk.


Journal of Immunology | 2003

Low Activation Threshold As a Mechanism for Ligand-Independent Signaling in Pre-T Cells

Mariëlle C. Haks; Stanley M. Belkowski; Maria Ciofani; Michele Rhodes; Juliette M. Lefebvre; Sébastien Trop; Patrice Hugo; Juan Carlos Zúñiga-Pflücker; David L. Wiest

Pre-TCR complexes are thought to signal in a ligand-independent manner because they are constitutively targeted to lipid rafts. We report that ligand-independent signaling is not a unique capability of the pre-TCR complex. Indeed, the TCRα subunit restores development of pTα-deficient thymocytes to the CD4+CD8+ stage even in the absence of conventional MHC class I and class II ligands. Moreover, we found that pre-TCR and αβTCR complexes exhibit no appreciable difference in their association with lipid rafts, suggesting that ligand-independence is a function of the CD4−CD8− (DN) thymocytes in which pre-TCR signaling occurs. In agreement, we found that only CD44−CD25+ DN thymocytes (DN3) enabled activation of extracellular signal-regulated kinases by the pre-TCR complex. DN thymocytes also exhibited a lower signaling threshold relative to CD4+CD8+ thymocytes, which was associated with both the markedly elevated lipid raft content of their plasma membranes and more robust capacitative Ca2+ entry. Taken together these data suggest that cell-autonomous, ligand-independent signaling is primarily a property of the thymocytes in which pre-TCR signaling occurs.


Journal of Immunology | 2005

Enforced Expression of Spi-B Reverses T Lineage Commitment and Blocks β-Selection

Juliette M. Lefebvre; Mariëlle C. Haks; Michael Carleton; Michele Rhodes; Gomathinayagam Sinnathamby; M. Celeste Simon; Laurence C. Eisenlohr; Lee Ann Garrett-Sinha; David L. Wiest

The molecular changes that restrict multipotent murine thymocytes to the T cell lineage and render them responsive to Ag receptor signals remain poorly understood. In this study, we report our analysis of the role of the Ets transcription factor, Spi-B, in this process. Spi-B expression is acutely induced coincident with T cell lineage commitment at the CD4−CD8−CD44−CD25+ (DN3) stage of thymocyte development and is then down-regulated as thymocytes respond to pre-TCR signals and develop beyond the β-selection checkpoint to the CD4−CD8−CD44−CD25− (DN4) stage. We found that dysregulation of Spi-B expression in DN3 thymocytes resulted in a dose-dependent perturbation of thymocyte development. Indeed, DN3 thymocytes expressing approximately five times the endogenous level of Spi-B were arrested at the β-selection checkpoint, due to impaired induction of Egr proteins, which are important molecular effectors of the β-selection checkpoint. T lineage-committed DN3 thymocytes expressing even higher levels of Spi-B were diverted to the dendritic cell lineage. Thus, we demonstrate that the prescribed modulation of Spi-B expression is important for T lineage commitment and differentiation beyond the β-selection checkpoint; and we provide insight into the mechanism underlying perturbation of development when that expression pattern is disrupted.


Developmental Cell | 2013

Control of Hematopoietic Stem Cell Emergence by Antagonistic Functions of Ribosomal Protein Paralogs

Yong Zhang; Anne-Cécile E. Duc; Shuyun Rao; Xiaoli Sun; Alison N. Bilbee; Michele Rhodes; Qin Li; Dietmar J. Kappes; Jennifer Rhodes; David L. Wiest

It remains controversial whether the highly homologous ribosomal protein (RP) paralogs found in lower eukaryotes have distinct functions and this has not been explored in vertebrates. Here we demonstrate that despite ubiquitous expression, the RP paralogs, Rpl22 and Rpl22-like1 (Rpl22l1) play essential, distinct, and antagonistic roles in hematopoietic development. Knockdown of Rpl22 in zebrafish embryos selectively blocks the development of T lineage progenitors after they have seeded the thymus. In contrast, knockdown of the Rpl22 paralog, Rpl22l1, impairs the emergence of hematopoietic stem cells (HSC) in the aorta-gonad-mesonephros by abrogating Smad1 expression and the consequent induction of essential transcriptional regulator, Runx1. Indeed, despite the ability of both paralogs to bind smad1 RNA, Rpl22 and Rpl22l1 have opposing effects on Smad1 expression. Accordingly, circumstances that tip the balance of these paralogs in favor of Rpl22 (e.g., Rpl22l1 knockdown or Rpl22 overexpression) result in repression of Smad1 and blockade of HSC emergence.


Journal of Immunology | 2000

Competitive Displacement of pTα by TCR-α During TCR Assembly Prevents Surface Coexpression of Pre-TCR and αβ TCR

Sébastien Trop; Michele Rhodes; David L. Wiest; Patrice Hugo; Juan Carlos Zúñiga-Pflücker

During αβ T cell development, CD4−CD8− thymocytes first express pre-TCR (pTα/TCR-β) before their differentiation to the CD4+CD8+ stage. Positive selection of self-tolerant T cells is then determined by the αβ TCR expressed on CD4+CD8+ thymocytes. Conceivably, an overlap in surface expression of these two receptors would interfere with the delicate balance of thymic selection. Therefore, a mechanism ensuring the sequential expression of pre-TCR and TCR must function during thymocyte development. In support of this notion, we demonstrate that expression of TCR-α by immature thymocytes terminates the surface expression of pre-TCR. Our results reveal that expression of TCR-α precludes the formation of pTα/TCR-β dimers within the endoplasmic reticulum, leading to the displacement of pre-TCR from the cell surface. These findings illustrate a novel posttranslational mechanism for the regulation of pre-TCR expression, which may ensure that αβ TCR expression on thymocytes undergoing selection is not compromised by the expression of pre-TCR.


Journal of Immunology | 2008

Egr2 Is Required for Bcl-2 Induction during Positive Selection

Jens-Peter Holst Lauritsen; Sridevi Kurella; Sang-Yun Lee; Juliette M. Lefebvre; Michele Rhodes; José Alberola-Ila; David L. Wiest

The repertoire of TCR specificities is established by a selection process in the thymus, during which precursor survival and maturation is dictated by the nature of the TCR signals. The differences in signals that determine whether precursors will survive and mature or be induced to die remain poorly understood. Among the molecular effectors involved in executing the differentiation process initiated by TCR-ligand interactions is a family of Zn-finger transcription factors termed early growth response genes (Egr). Indeed, ablation of the Egr1 gene impairs ligand-induced maturation (positive selection) but not ligand-induced deletion (negative selection). The partial impairment of positive selection by Egr1 deficiency is not enhanced by simultaneous deletion of another Egr family member, Egr3. Accordingly, we asked whether this results from compensation by another family member, Egr2. In this manuscript, we demonstrate that deletion of Egr2 impairs positive selection of both CD4 and CD8 single-positive thymocytes. Interestingly, many of the genes involved in positive selection and T cell differentiation are up-regulated normally in the Egr2-deficient thymocytes. However, Bcl-2 up-regulation is not sustained during late stages of positive selection. This defect is at least partially responsible for the developmental blockade in Egr2-deficient thymocytes, as enforced expression of Bcl-2 rescues T cell development in Egr2−/− thymocytes. Taken together, these data suggest that Egr2 plays a central role in the up-regulation of the survival molecule Bcl-2 during positive selection.


Journal of Immunology | 2004

Architectural Changes in the TCR:CD3 Complex Induced by MHC:Peptide Ligation

Nicole L. La Gruta; Haiyan Liu; Smaroula Dilioglou; Michele Rhodes; David L. Wiest; Dario A. A. Vignali

A hallmark of T cell activation is the ligation-induced down-modulation of the TCR:CD3 complex. However, little is known about the molecular events that drive this process. The CD3 ζ-chain has been shown to play a unique role in regulating the assembly, transport, and cell surface expression of the TCR:CD3 complex. In this study we have investigated the relationship between CD3ζ and the TCRαβCD3εδγ complex after ligation by MHC:peptide complexes. Our results show that there is a significant increase in free surface CD3ζ, which is not associated with the TCR:CD3 complex, after T cell stimulation. This may reflect dissociation of CD3ζ from the TCRαβCD3εδγ complex or transport of intracellular CD3ζ directly to the cell surface. We also show that MHC:peptide ligation also results in exposure of the TCR-associated CD3ζ NH2 terminus, which is ordinarily buried in the complex. These observations appears to be dependent on Src family protein tyrosine kinases, which are known to be critical for efficient T cell activation. These data suggest a mechanism by which ligated TCR may be differentiated from unligated TCR and selectively down-modulated.


Journal of Experimental Medicine | 2016

Mutations in STN1 cause Coats plus syndrome and are associated with genomic and telomere defects

Amos J. Simon; Atar Lev; Yong Zhang; Batia Weiss; Anna Rylova; Eran Eyal; Nitzan Kol; Ortal Barel; Keren Cesarkas; Michalle Soudack; Noa Greenberg-Kushnir; Michele Rhodes; David L. Wiest; Ginette Schiby; Iris Barshack; Shulamit Katz; Elon Pras; Hana Poran; Haike Reznik-Wolf; Elena Ribakovsky; Carlos Simon; Wadi Hazou; Yechezkel Sidi; Avishay Lahad; Hagar Katzir; Shira Sagie; Haifa A. Aqeilan; Galina Glousker; Ninette Amariglio; Yehuda Tzfati

Somech and colleagues identify two new mutations in STN1 that causes Coats plus syndrome and telomere abnormalities in human, recapitulated in a zebra fish model.

Collaboration


Dive into the Michele Rhodes's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shuyun Rao

Fox Chase Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sang-Yun Lee

Fox Chase Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yong Zhang

Fox Chase Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerard P. Zambetti

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge