Network


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

Hotspot


Dive into the research topics where Richard N. Hanna is active.

Publication


Featured researches published by Richard N. Hanna.


Cell | 2013

Nr4a1-Dependent Ly6C(low) Monocytes Monitor Endothelial Cells and Orchestrate Their Disposal

Leo M. Carlin; Efstathios G. Stamatiades; Cédric Auffray; Richard N. Hanna; Leanne Glover; Gema Vizcay-Barrena; Catherine C. Hedrick; H. Terence Cook; Sandra S. Diebold; Frederic Geissmann

Summary The functions of Nr4a1-dependent Ly6Clow monocytes remain enigmatic. We show that they are enriched within capillaries and scavenge microparticles from their lumenal side in a steady state. In the kidney cortex, perturbation of homeostasis by a TLR7-dependent nucleic acid “danger” signal, which may signify viral infection or local cell death, triggers Gαi-dependent intravascular retention of Ly6Clow monocytes by the endothelium. Then, monocytes recruit neutrophils in a TLR7-dependent manner to mediate focal necrosis of endothelial cells, whereas the monocytes remove cellular debris. Prevention of Ly6Clow monocyte development, crawling, or retention in Nr4a1−/−, Itgal−/−, and Tlr7host−/−BM+/+ and Cx3cr1−/− mice, respectively, abolished neutrophil recruitment and endothelial killing. Prevention of neutrophil recruitment in Tlr7host+/+BM−/− mice or by neutrophil depletion also abolished endothelial cell necrosis. Therefore, Ly6Clow monocytes are intravascular housekeepers that orchestrate the necrosis by neutrophils of endothelial cells that signal a local threat sensed via TLR7 followed by the in situ phagocytosis of cellular debris.


Circulation Research | 2012

NR4A1 (Nur77) Deletion Polarizes Macrophages Toward an Inflammatory Phenotype and Increases Atherosclerosis

Richard N. Hanna; Iftach Shaked; Harper Hubbeling; Jennifer A. Punt; Runpei Wu; Claudia Zaugg; Hong Pei; Frederic Geissmann; Klaus Ley; Catherine C. Hedrick

Rationale: NR4A1 (Nur77) is a nuclear receptor that is expressed in macrophages and within atherosclerotic lesions, yet its function in atherosclerosis is unknown. Objective: Nur77 regulates the development of monocytes, particularly patrolling Ly6C− monocytes that may be involved in resolution of inflammation. We sought to determine how absence of nuclear receptor subfamily 4, group A, member 1 (NR4A1) in hematopoietic cells affected atherosclerosis development. Methods and Results: Nur77−/− chimeric mice on a Ldlr−/− background showed a 3-fold increase in atherosclerosis development when fed a Western diet for 20 weeks, despite having a drastic reduction in Ly6C− patrolling monocytes. In a second model, mice deficient in both Nur77 and ApoE (ApoE−/−Nur77−/−) also showed increased atherosclerosis after 11 weeks of Western diet. Atherosclerosis was associated with a significant change in macrophage polarization toward a proinflammatory phenotype, with high expression of tumor necrosis factor-&agr; and nitric oxide and low expression of Arginase-I. Moreover, we found increased expression of toll-like receptor 4 mRNA and protein in Nur77−/− macrophages as well as increased phosphorylation of the p65 subunit of NF&kgr;B. Inhibition of NF&kgr;B activity blocked excess activation of Nur77−/− macrophages. Conclusions: We conclude that the absence of Nur77 in monocytes and macrophages results in enhanced toll-like receptor signaling and polarization of macrophages toward a proinflammatory M1 phenotype. Despite having fewer monocytes, Nur77−/− mice developed significant atherosclerosis when fed a Western diet. These studies indicate that Nur77 is a novel target for modulating the inflammatory phenotype of monocytes and macrophages and may be important for regulation of atherogenesis.


Science | 2015

Patrolling monocytes control tumor metastasis to the lung

Richard N. Hanna; Caglar Cekic; Duygu Sag; Robert Tacke; Graham D. Thomas; Heba Nowyhed; Nicole Rasquinha; Sara McArdle; Runpei Wu; Esther Peluso; Daniel Metzger; Hiroshi Ichinose; Iftach Shaked; Grzegorz Chodaczek; Subhra K. Biswas; Catherine C. Hedrick

Monocytes block tumor access to the lung Metastatic cancer is especially hard to treat. In order to find potential new therapeutic targets, scientists are trying to understand the cellular events that promote or prevent metastasis. Hanna et al. now report a role for patrolling monocytes in blocking tumor metastasis to the lungs in mice. Tumors in mice engineered to lack patrolling monocytes showed increased metastasis to the lung but not to other tissues. Patrolling monocytes resided in the microvasculature of the lung, where they engulfed tumor material, which may explain how these cells prevent tumors from colonizing the lung. Science, this issue p. 985 An immune cell subset called patrolling monocytes prevents tumor lung metastasis in mice. The immune system plays an important role in regulating tumor growth and metastasis. Classical monocytes promote tumorigenesis and cancer metastasis, but how nonclassical “patrolling” monocytes (PMo) interact with tumors is unknown. Here we show that PMo are enriched in the microvasculature of the lung and reduce tumor metastasis to lung in multiple mouse metastatic tumor models. Nr4a1-deficient mice, which specifically lack PMo, showed increased lung metastasis in vivo. Transfer of Nr4a1-proficient PMo into Nr4a1-deficient mice prevented tumor invasion in the lung. PMo established early interactions with metastasizing tumor cells, scavenged tumor material from the lung vasculature, and promoted natural killer cell recruitment and activation. Thus, PMo contribute to cancer immunosurveillance and may be targets for cancer immunotherapy.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2015

Nonclassical Patrolling Monocyte Function in the Vasculature

Graham D. Thomas; Robert Tacke; Catherine C. Hedrick; Richard N. Hanna

Nonclassical patrolling monocytes are characterized by their unique ability to actively patrol the vascular endothelium under homeostatic and inflammatory conditions. Patrolling monocyte subsets (CX3CR1(high)Ly6C(-) in mouse and CX3CR1(high)CD14(dim)CD16(+) in humans) are distinct from the classical monocyte subsets (CCR2(high)Ly6C(+) in mouse and CCR2(high)CD14(+)CD16(-) in humans) and exhibit unique functions in the vasculature and inflammatory disease. Patrolling monocytes function in several disease settings to remove damaged cells and debris from the vasculature and have been associated with wound healing and the resolution of inflammation in damaged tissues. This review highlights the unique functions of these patrolling monocytes in the vasculature and during inflammation.


Circulation | 2013

Molecular Imaging of the Paracrine Proangiogenic Effects of Progenitor Cell Therapy in Limb Ischemia

Jae Choon Ryu; Brian P. Davidson; Aris Xie; Yue Qi; Daogang Zha; J. Todd Belcik; Evan S. Caplan; Juliana Woda; Catherine C. Hedrick; Richard N. Hanna; Nicholas Lehman; Yan Zhao; Anthony E. Ting; Jonathan R. Lindner

Background— Stem cells are thought to enhance vascular remodeling in ischemic tissue in part through paracrine effects. Using molecular imaging, we tested the hypothesis that treatment of limb ischemia with multipotential adult progenitor cells (MAPCs) promotes recovery of blood flow through the recruitment of proangiogenic monocytes. Methods and Results— Hind-limb ischemia was produced in mice by iliac artery ligation, and MAPCs were administered intramuscularly on day 1. Optical imaging of luciferase-transfected MAPCs indicated that cells survived for 1 week. Contrast-enhanced ultrasound on days 3, 7, and 21 showed a more complete recovery of blood flow and greater expansion of microvascular blood volume in MAPC-treated mice than in controls. Fluorescent microangiography demonstrated more complete distribution of flow to microvascular units in MAPC-treated mice. On ultrasound molecular imaging, expression of endothelial P-selectin and intravascular recruitment of CX3CR-1-positive monocytes were significantly higher in MAPC-treated mice than in the control groups at days 3 and 7 after arterial ligation. Muscle immunohistology showed a >10-fold-greater infiltration of monocytes in MAPC-treated than control-treated ischemic limbs at all time points. Intravital microscopy of ischemic or tumor necrosis factor-&agr;–treated cremaster muscle demonstrated that MAPCs migrate to perimicrovascular locations and potentiate selectin-dependent leukocyte rolling. In vitro migration of human CD14+ monocytes was 10-fold greater in response to MAPC-conditioned than basal media. Conclusions— In limb ischemia, MAPCs stimulate the recruitment of proangiogenic monocytes through endothelial activation and enhanced chemotaxis. These responses are sustained beyond the MAPC lifespan, suggesting that paracrine effects promote flow recovery by rebalancing the immune response toward a more regenerative phenotype.


Current Opinion in Lipidology | 2013

NR4A Nuclear Receptors in Immunity and Atherosclerosis

Anouk A.J. Hamers; Richard N. Hanna; Heba Nowyhed; Catherine C. Hedrick; Carlie J.M. de Vries

Purpose of review To understand chronic inflammatory diseases such as atherosclerosis, we require in-depth knowledge on immune-cell differentiation, function of specific immune-cell subsets and endothelial cell-mediated extravasation. In this review, we summarize a number of very recent observations on the pivotal function of NR4A nuclear receptors in immunity and atherosclerosis. Recent findings NR4A nuclear receptors are involved in negative selection of thymocytes, Treg differentiation and the development of Ly6C- monocytes. Nur77 and Nurr1 attenuate atherosclerosis in mice whereas NOR-1 aggravates vascular lesion formation. Summary These exciting, novel insights on the function of NR4A nuclear receptors in immunity, vascular cells and atherosclerosis will initiate a plethora of studies to understand the underlying molecular mechanisms, which will culminate in the identification of novel NR4A targets to modulate chronic inflammatory disease.


Nature Immunology | 2015

Transcription factor Nr4a1 couples sympathetic and inflammatory cues in CNS-recruited macrophages to limit neuroinflammation

Iftach Shaked; Richard N. Hanna; Helena Shaked; Grzegorz Chodaczek; Heba Nowyhed; George Tweet; Robert Tacke; Alp Bugra Basat; Zbigniew Mikulski; Susan Togher; Jacqueline Miller; Amy Blatchley; Shahram Salek-Ardakani; Martin Darvas; Minna U. Kaikkonen; Graham D. Thomas; Sonia Lai-Wing-Sun; Ayman Rezk; Amit Bar-Or; Christopher K. Glass; Hozefa S. Bandukwala; Catherine C. Hedrick

The molecular mechanisms that link the sympathetic stress response and inflammation remain obscure. Here we found that the transcription factor Nr4a1 regulated the production of norepinephrine (NE) in macrophages and thereby limited experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. Lack of Nr4a1 in myeloid cells led to enhanced NE production, accelerated infiltration of leukocytes into the central nervous system (CNS) and disease exacerbation in vivo. In contrast, myeloid-specific deletion of tyrosine hydroxylase (TH), the rate-limiting enzyme in catecholamine biosynthesis, protected mice against EAE. Furthermore, we found that Nr4a1 repressed autocrine NE production in macrophages by recruiting the corepressor CoREST to the Th promoter. Our data reveal a new role for macrophages in neuroinflammation and identify Nr4a1 as a key regulator of catecholamine production by macrophages.


PLOS ONE | 2013

Increased cholesterol content in gammadelta (γδ) T lymphocytes differentially regulates their activation.

Hsin-Yuan Cheng; Runpei Wu; Abraham K. Gebre; Richard N. Hanna; Daniel J. Smith; John S. Parks; Klaus Ley; Catherine C. Hedrick

Gammadelta (γδ) T lymphocytes respond quickly upon antigen encounter to produce a cytokine response. In this study, we sought to understand how functions of γδ T cells are differentially regulated compared to αβ T cells. We found that cholesterol, an integral component of the plasma membrane and a regulator of TCR signaling, is increased in γδ T cells compared to αβ T cells, and modulates their function. Higher levels of activation markers, and increased lipid raft content in γδ cells suggest that γδ T cells are more activated. Cholesterol depletion effectively decreased lipid raft formation and activation of γδ T cells, indicating that increased cholesterol content contributes to the hyper-activated phenotype of γδ T cells, possibly through enhanced clustering of TCR signals in lipid rafts. TCR stimulation assays and western blotting revealed that instead of a lower TCR threshold, enhanced TCR signaling through ERK1/2 activation is likely the cause for high cholesterol-induced rapid activation and proliferation in γδ T cells. Our data indicate that cholesterol metabolism is differentially regulated in γδ T cells. The high intracellular cholesterol content leads to enhanced TCR signaling and increases activation and proliferation of γδ T cells.


Scientific Reports | 2015

The transcription factor NR4A1 is essential for the development of a novel macrophage subset in the thymus

Robert Tacke; Ingo Hilgendorf; Hannah Garner; Claire Waterborg; Kiwon Park; Heba Nowyhed; Richard N. Hanna; Runpei Wu; Filip K. Swirski; Frederic Geissmann; Catherine C. Hedrick

Tissue macrophages function to maintain homeostasis and regulate immune responses. While tissue macrophages derive from one of a small number of progenitor programs, the transcriptional requirements for site-specific macrophage subset development are more complex. We have identified a new tissue macrophage subset in the thymus and have discovered that its development is dependent on transcription factor NR4A1. Functionally, we find that NR4A1-dependent macrophages are critically important for clearance of apoptotic thymocytes. These macrophages are largely reduced or absent in mice lacking NR4A1, and Nr4a1-deficient mice have impaired thymocyte engulfment and clearance. Thus, NR4A1 functions as a master transcription factor for the development of this novel thymus-specific macrophage subset.


Nature Medicine | 2014

Stressing out stem cells: linking stress and hematopoiesis in cardiovascular disease

Richard N. Hanna; Catherine C. Hedrick

Stress has long been thought to be a major contributing factor to cardiovascular disease, although little is known about the underlying cellular mechanisms. A new study in humans and mice suggests that chronic stress promotes hematopoietic stem cell proliferation in bone marrow, leading to increased leukocyte production, circulation and recruitment to the vasculature (pages 754–758).

Collaboration


Dive into the Richard N. Hanna's collaboration.

Top Co-Authors

Avatar

Catherine C. Hedrick

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Graham D. Thomas

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Iftach Shaked

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Frederic Geissmann

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Heba Nowyhed

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Klaus Ley

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Robert Tacke

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Runpei Wu

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Grzegorz Chodaczek

La Jolla Institute for Allergy and Immunology

View shared research outputs
Researchain Logo
Decentralizing Knowledge