Network


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

Hotspot


Dive into the research topics where Mark C. Udey is active.

Publication


Featured researches published by Mark C. Udey.


Journal of Clinical Investigation | 2005

Characterization and isolation of stem cell–enriched human hair follicle bulge cells

Manabu Ohyama; Atsushi Terunuma; Christine L. Tock; Michael F. Radonovich; Cynthia A. Pise-Masison; Steven B. Hopping; John N. Brady; Mark C. Udey; Jonathan C. Vogel

The human hair follicle bulge is an important niche for keratinocyte stem cells (KSCs). Elucidation of human bulge cell biology could be facilitated by analysis of global gene expression profiles and identification of unique cell-surface markers. The lack of distinctive bulge morphology in human hair follicles has hampered studies of bulge cells and KSCs. In this study, we determined the distribution of label-retaining cells to define the human anagen bulge. Using navigated laser capture microdissection, bulge cells and outer root sheath cells from other follicle regions were obtained and analyzed with cDNA microarrays. Gene transcripts encoding inhibitors of WNT and activin/bone morphogenic protein signaling were overrepresented in the bulge, while genes responsible for cell proliferation were underrepresented, consistent with the existence of quiescent noncycling KSCs in anagen follicles. Positive markers for bulge cells included CD200, PHLDA1, follistatin, and frizzled homolog 1, while CD24, CD34, CD71, and CD146 were preferentially expressed by non-bulge keratinocytes. Importantly, CD200+ cells (CD200hiCD24loCD34loCD71loCD146lo) obtained from hair follicle suspensions demonstrated high colony-forming efficiency in clonogenic assays, indicating successful enrichment of living human bulge stem cells. The stem cell behavior of enriched bulge cells and their utility for gene therapy and hair regeneration will need to be assessed in in vivo assays.


Journal of Immunology | 2002

CD8+ T Cells Are Required for Primary Immunity in C57BL/6 Mice Following Low-Dose, Intradermal Challenge with Leishmania major

Yasmine Belkaid; Esther von Stebut; Susana Mendez; Rosalia Lira; Elisabet Caler; Sylvie Bertholet; Mark C. Udey; David L. Sacks

Standard murine models of cutaneous leishmaniasis, involving s.c. inoculation of large numbers of Leishmania major promastigotes, have not supported an essential role for CD8+ T cells in the control of primary infection. Recently, a L. major model combining two main features of natural transmission, low parasite dose and inoculation into a dermal site, has been established in resistant C57BL/6 mice. In the present studies, C57BL/6 mice with CD8+ T cell deficiencies, including CD8−/− and CD8-depleted mice, failed to control the growth of L. major following inoculation of 100 metacyclic promastigotes into the ear dermis. The resulting dermal pathology was minor and delayed. Lesion formation in wild-type mice was coincident with the killing of parasites in the inoculation site. Both events were associated with the accumulation of CD8+ T lymphocytes in the skin and with the capacity of CD8+ T cells recovered from draining lymph nodes or infected dermis to release IFN-γ following coculture with infected dendritic cells. Reconstitution of resistance to L. major in RAG−/− mice using T cells from naive donors was optimal when both CD4+ and CD8+ T cells were transferred. Primed CD8+ T lymphocytes obtained from C57BL/6 mice during the acute stage of infection were able to mediate both pathology and immunity when transferred alone. The low dose, intradermal challenge model reveals that CD8+ T cells play an essential role in both pathogenesis of and immunity to primary infection with L. major in the skin.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Murine epidermal Langerhans cells and langerin-expressing dermal dendritic cells are unrelated and exhibit distinct functions

Keisuke Nagao; Florent Ginhoux; Wolfgang W. Leitner; Sei-ichiro Motegi; Clare L. Bennett; Björn E. Clausen; Miriam Merad; Mark C. Udey

A new langerin+ DC subset has recently been identified in murine dermis (langerin+ dDC), but the lineage and functional relationships between these cells and langerin+ epidermal Langerhans cells (LC) are incompletely characterized. Selective expression of the cell adhesion molecule EpCAM by LC allowed viable LC to be easily distinguished from langerin+ dDC in skin and lymphoid tissue and ex vivo as well. Differential expression of EpCAM and langerin revealed the presence of at least 3 distinct skin DC subsets. We determined that LC and langerin+ dDC exhibit different migratory capabilities in vitro and repopulate distinct anatomic compartments in skin at different rates after conditional depletion in vivo. Langerin+ dDC, in contrast to LC, did not require TGFβ1 for development. Carefully timed gene gun immunization studies designed to take advantage of the distinct repopulation kinetics of langerin+ dDC and LC revealed that langerin+ dDC were required for optimal production of β-galactosidase-specific IgG2a/c and IgG2b in the acute phase. In contrast, immunization via LC-deficient skin resulted in persistent and strikingly reduced IgG1 and enhanced IgG2a Ab production. Our data support the concepts that LC and langerin+ dDC represent distinct DC subsets that have specialized functions and that LC are important immunoregulatory cells. The presence of at least 3 functionally distinct skin DC subsets may have particular relevance for vaccines that are administered epicutaneously.


Journal of Experimental Medicine | 2006

Uptake of Leishmania major by dendritic cells is mediated by Fcγ receptors and facilitates acquisition of protective immunity

Florian Woelbing; Susanna Lopez Kostka; Katharina Moelle; Yasmine Belkaid; Cord Sunderkoetter; Sjef Verbeek; Ari Waisman; Axel P. Nigg; Juergen Knop; Mark C. Udey; Esther von Stebut

Uptake of Leishmania major by dendritic cells (DCs) results in activation and interleukin (IL)-12 release. Infected DCs efficiently stimulate CD4− and CD8− T cells and vaccinate against leishmaniasis. In contrast, complement receptor 3–dependent phagocytosis of L. major by macrophages (MΦ) leads exclusively to MHC class II–restricted antigen presentation to primed, but not naive, T cells, and no IL-12 production. Herein, we demonstrate that uptake of L. major by DCs required parasite-reactive immunoglobulin (Ig)G and involved FcγRI and FcγRIII. In vivo, DC infiltration of L. major–infected skin lesions coincided with the appearance of antibodies in sera. Skin of infected B cell–deficient mice and Fcγ−/− mice contained fewer parasite-infected DCs in vivo. Infected B cell–deficient mice as well as Fcγ−/− mice (all on the C57BL/6 background) showed similarly increased disease susceptibility as assessed by lesion volumes and parasite burdens. The B cell–deficient mice displayed impaired T cell priming and dramatically reduced IFN-γ production, and these deficits were normalized by infection with IgG-opsonized parasites. These data demonstrate that DC and MΦ use different receptors to recognize and ingest L. major with different outcomes, and indicate that B cell–derived, parasite-reactive IgG and DC FcγRI and FcγRIII are essential for optimal development of protective immunity.


Science | 2008

Del-1, an Endogenous Leukocyte-Endothelial Adhesion Inhibitor, Limits Inflammatory Cell Recruitment

Eun Young Choi; Emmanouil Chavakis; Marcus Czabanka; Harald Langer; Line Fraemohs; Matina Economopoulou; Ramendra K. Kundu; Alessia Orlandi; Ying Yi Zheng; DaRue A. Prieto; Christie M. Ballantyne; Stephanie L. Constant; William C. Aird; Thalia Papayannopoulou; Carl G. Gahmberg; Mark C. Udey; Peter Vajkoczy; Thomas Quertermous; Stefanie Dimmeler; Christian Weber; Triantafyllos Chavakis

Leukocyte recruitment to sites of infection or inflammation requires multiple adhesive events. Although numerous players promoting leukocyte-endothelial interactions have been characterized, functionally important endogenous inhibitors of leukocyte adhesion have not been identified. Here we describe the endothelially derived secreted molecule Del-1 (developmental endothelial locus–1) as an anti-adhesive factor that interferes with the integrin LFA-1–dependent leukocyte-endothelial adhesion. Endothelial Del-1 deficiency increased LFA-1–dependent leukocyte adhesion in vitro and in vivo. Del-1–/– mice displayed significantly higher neutrophil accumulation in lipopolysaccharide-induced lung inflammation in vivo, which was reversed in Del-1/LFA-1 double-deficient mice. Thus, Del-1 is an endogenous inhibitor of inflammatory cell recruitment and could provide a basis for targeting leukocyte-endothelial interactions in disease.


Nature Immunology | 2012

Stress-induced production of chemokines by hair follicles regulates the trafficking of dendritic cells in skin

Keisuke Nagao; Tetsuro Kobayashi; Kazuyo Moro; Manabu Ohyama; Takeya Adachi; Daniela Y. Kitashima; Satoshi Ueha; Keisuke Horiuchi; Hideaki Tanizaki; Kenji Kabashima; Akiharu Kubo; Young Hun Cho; Bjarn E. Clausen; Kouji Matsushima; Makoto Suematsu; Glaucia C. Furtado; Sergio A. Lira; Joshua M. Farber; Mark C. Udey; Masayuki Amagai

Langerhans cells (LCs) are epidermal dendritic cells with incompletely understood origins that associate with hair follicles for unknown reasons. Here we show that in response to external stress, mouse hair follicles recruited Gr-1hi monocyte-derived precursors of LCs whose epidermal entry was dependent on the chemokine receptors CCR2 and CCR6, whereas the chemokine receptor CCR8 inhibited the recruitment of LCs. Distinct hair-follicle regions had differences in their expression of ligands for CCR2 and CCR6. The isthmus expressed the chemokine CCL2; the infundibulum expressed the chemokine CCL20; and keratinocytes in the bulge produced the chemokine CCL8, which is the ligand for CCR8. Thus, distinct hair-follicle keratinocyte subpopulations promoted or inhibited repopulation with LCs via differences in chemokine production, a feature also noted in humans. Pre-LCs failed to enter hairless skin in mice or humans, which establishes hair follicles as portals for LCs.


Journal of Immunology | 2002

Dendritic cells transduced with protein antigens induce cytotoxic lymphocytes and elicit antitumor immunity.

Naotaka Shibagaki; Mark C. Udey

Dendritic cell (DC)-based vaccines are being developed for treatment of patients with cancer, in part because DC are potent inducers of CD8+ CTL. DC MHC class I:antigenic peptide complexes that are required for CTL elicitation are most often generated by incubating DC with peptides or by transfecting (or transducing) DC with cDNAs (or viral vectors) that encode protein Ags. The former approach is feasible when MHC class I Ags and relevant peptides are known. The latter approach may be hampered by inefficient DC transfection (transduction) and/or difficulties associated with preparation and use of viral vectors. Herein we demonstrate that a bacterial recombinant model tumor-associated Ag (OVA) that contains the HIV TAT protein transduction domain (PTD) was readily engineered and purified, efficiently transduced murine lymphocytes and DC, and was processed by proteasomes for MHC class I-restricted presentation to CTL. In addition, PTD-containing rOVA was processed and presented by DC to CD4 T cells as efficiently as native OVA or rOVA lacking the PTD. PTD-OVA-transduced DC induced CTL in vivo in a Th cell-independent fashion and vaccinated against OVA-expressing tumors. In contrast, rOVA lacking the PTD did not enter the DC MHC class I presentation pathway and DC treated with this protein did not prime OVA-specific CTL in vivo. Treatment of mice harboring clinically apparent OVA-expressing tumors with PTD-OVA-transduced DC resulted in tumor regression in some animals. This straightforward vaccination strategy may translate into DC-based treatments for patients with cancer and other serious diseases.


Journal of Immunology | 2009

IL-17 promotes progression of cutaneous leishmaniasis in susceptible mice

Susanna Lopez Kostka; Stephanie Dinges; Klaus Griewank; Yoichiro Iwakura; Mark C. Udey; Esther von Stebut

Resistance to leishmaniasis in C57BL/6 mice depends on Th1/Tc1 cells. BALB/c mice preferentially develop Th2 immunity and succumb to infection. We now assessed the role of IL-17 in cutaneous leishmaniasis. During the course of Leishmania major infection, BALB/c CD4 cells and neutrophils produced increased amounts of IL-17 as compared with cells from C57BL/6 mice. This increase was associated with significantly increased IL-23 release from L. major-infected BALB/c dendritic cells (DC), whereas IL-6 and TGF-β1 production by BALB/c and C57BL/6 DC were comparable. Interestingly, lesion sizes in infected IL-17-deficient BALB/c mice were dramatically smaller and failed to progress as compared with those in control mice. Similar amounts of IL-4, IL-10, and IFN-γ were produced by T cells from IL-17-deficient mice and control mice consistent with development of Th2-predominant immunity in all animals. Improved disease outcome was associated with decreased CXCL2-accumulation in lesion sites and decreased neutrophil immigration into lesions of infected IL-17-deficient mice confirming prior observations that enhanced neutrophil recruitment contributes to disease susceptibility in BALB/c mice. This study excludes an important facilitating role for IL-17 in Th1/Th2 development in L. major-infected BALB/c mice, and suggests that IL-23 production by L. major-infected DC maintains IL-17+ cells that influence disease progression via regulation of neutrophil recruitment.


European Journal of Immunology | 2000

Leishmania major‐infected murine Langerhans cell‐like dendritic cells from susceptible mice release IL‐12 after infection and vaccinate against experimental cutaneous Leishmaniasis

Esther von Stebut; Yasmine Belkaid; Bai V. Nguyen; Melissa Cushing; David L. Sacks; Mark C. Udey

Leishmania major‐infected C57BL / 6 skin‐dendritic cells (DC) are activated and release cytokines (including IL‐12 p70), and likely initiate protective Th1 immunity in vivo (von Stebut, E. et al., J. Exp. Med. 188: 1547 – 1552). To characterize differences in DC function in mice that are genetically susceptible (BALB / c) and resistant (C57BL / 6) to cutaneous leishmaniasis, we analyzed the effects of L. major on Langerhans cell‐like, fetal skin‐derived DC (FSDDC) from both strains. BALB / c‐ and C57BL / 6‐FSDDC ingested similar numbers of amastigotes, but did not ingest metacyclic promastigotes. Like C57BL / 6‐FSDDC, infection of BALB / c‐FSDDC led to up‐regulation of MHC class I and II antigens, CD40, CD54, and CD86 within 18 h. L. major‐induced BALB / c DC activation also led to the release of TNF‐α, IL‐6 and IL‐12 p40 into 18‐h supernatants. Infected BALB / c‐ and C57BL / 6‐DC both released small amounts of IL‐12 p70 within 72 h. Additional stimulation with IFN‐γ and / or anti‐CD40 induced the release of more IL‐12 p70 from infected BALB / c‐DC than C57BL / 6‐DC. Co‐culture of control or infected BALB / c‐ and C57BL / 6‐DC with naive syngeneic CD4+ T cells and soluble anti‐CD3 resulted in mixed, IFN‐γ‐predominant responses after restimulation with immobilized anti‐CD3. Finally, syngeneic L. major‐infected DC effectively vaccinated BALB / c mice against cutaneous leishmaniasis. Genetic susceptibility to L. major that results from induction of Th2 predominant immune responses after infection does not appear to reflect failure of skin DC to internalize or respond to parasites, or the inability of BALB / c T cells to mount a Th1 response to DC‐associated Leishmania antigens.


Journal of Clinical Investigation | 1997

A role for TGFbeta1 in langerhans cell biology. Further characterization of the epidermal Langerhans cell defect in TGFbeta1 null mice.

Teresa A. Borkowski; John J. Letterio; Crystal L. Mackall; Atsushi Saitoh; Xiao-Jing Wang; Dennis R. Roop; Ronald E. Gress; Mark C. Udey

Previous studies of TGFbeta1 null (-/-) mice indicated that the epidermis was devoid of Langerhans cells (LC) and that the LC deficiency was not secondary to the inflammation that is the dominant feature of the -/- phenotype (Borkowski, T.A., J.J. Letterio, A.G. Farr, and M.C. Udey. 1996. J. Exp. Med. 184:2417-2422). Herein, we demonstrate that dendritic cells could be expanded from the bone marrow of -/- mice and littermate controls. Bone marrow from -/- mice also gave rise to LC after transfer into lethally irradiated recipients. Thus, the LC defect in TGFbeta1 null mice does not result from an absolute deficiency in bone marrow precursors, and paracrine TGFbeta1 production is sufficient for LC development. Several approaches were used to assess the suitability of -/- skin for LC localization. A survey revealed that although a number of cytokine mRNAs were expressed de novo, mRNAs encoding proinflammatory cytokines known to mobilize LC from epidermis (IL-1 and TNFalpha) were not strikingly overrepresented in -/- skin. In addition, bone marrow-derived LC populated full-thickness TGFbeta1 null skin after engraftment onto BALB/c nu/nu recipients. Finally, the skin of transgenic mice expressing a truncated loricrin promoter-driven dominant-negative TGFbeta type II receptor contained normal numbers of LC. Because TGFbeta1 signaling in these mice is disrupted only in keratinocytes and the keratinocyte hyperproliferative component of the TGFbeta1 -/- phenotype is reproduced, these results strongly suggest that the LC defect in TGFbeta1 null mice is not due to an epidermal abnormality but reflects a requirement of murine LC (or their precursors) for TGFbeta1.

Collaboration


Dive into the Mark C. Udey's collaboration.

Top Co-Authors

Avatar

Jonathan C. Vogel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yasmine Belkaid

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Teresa A. Borkowski

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Andrew G. Farr

University of Washington

View shared research outputs
Top Co-Authors

Avatar

David L. Sacks

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Maria I. Morasso

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge