Network


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

Hotspot


Dive into the research topics where Kazuyo Yamaji-Kegan is active.

Publication


Featured researches published by Kazuyo Yamaji-Kegan.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2009

Hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMα) induces the vascular and hemodynamic changes of pulmonary hypertension

Daniel J. Angelini; Qingning Su; Kazuyo Yamaji-Kegan; Chunling Fan; John Skinner; Hunter C. Champion; Michael T. Crow; Roger A. Johns

Pulmonary hypertension (PH) is a serious disease of multiple etiologies mediated by hypoxia, immune stimuli, and elevated pulmonary pressure that leads to vascular thickening and eventual right heart failure. In a chronic hypoxia model of PH, we previously reported the induction of a novel pleiotropic cytokine, hypoxia-induced mitogenic factor (HIMF), that exhibits mitogenic, vasculogenic, contractile, and chemokine properties during PH-associated vascular remodeling. To examine the role of HIMF in hypoxia-induced vascular remodeling, we performed in vivo knockdown of HIMF using short hairpin RNA directed at rat HIMF in the chronic hypoxia model of PH. Knockdown of HIMF partially blocked increases in mean pulmonary artery pressure, pulmonary vascular resistance, right heart hypertrophy, and vascular remodeling caused by chronic hypoxia. To demonstrate a direct role for HIMF in the mechanism of PH development, we performed HIMF-gene transfer into the lungs of rats using a HIMF-expressing adeno-associated virus (AAV). AAV-HIMF alone caused development of PH similar to that of chronic hypoxia with increased mean pulmonary artery pressure and pulmonary vascular resistance, right heart hypertrophy, and neomuscularization and thickening of small pulmonary arterioles. The findings suggest that HIMF represents a critical cytokine-like growth factor in the development of PH.


Journal of Immunology | 2010

Hypoxia-Induced Mitogenic Factor (HIMF/FIZZ1/RELMα) Increases Lung Inflammation and Activates Pulmonary Microvascular Endothelial Cells via an IL-4–Dependent Mechanism

Kazuyo Yamaji-Kegan; Qingning Su; Daniel J. Angelini; Allen C. Myers; Chris Cheadle; Roger A. Johns

Hypoxia-induced mitogenic factor (HIMF), also known as found in inflammatory zone 1 and resistin-like molecule α, belongs to a novel class of cysteine-rich secreted proteins. It exhibits mitogenic and chemotactic properties during pulmonary hypertension-associated vascular remodeling, as well as fibrogenic properties during pulmonary fibrosis. HIMF expression in the lung was reported to be regulated by Th2 cytokines (IL-4 and IL-13) via the transcription factor STAT6 pathway in a bleomycin-induced pulmonary fibrosis model. However, in this study, we found that in the hypoxia-induced pulmonary hypertension model, lung HIMF expression is increased in IL-4 and STAT6 knockout (KO) mice to the same degree as in wild-type (WT) mice, suggesting that induction of HIMF expression does not require Th2 regulation in this model. We also found that HIMF-induced proliferative activity, hypertrophy, collagen, and extracellular matrix deposition in the pulmonary arteries are significantly less in IL-4 KO mice than in WT mice. In addition, HIMF-induced production of angiogenic factors/chemokines, such as vascular endothelial growth factor, MCP-1, and stromal-derived factor-1, in the lung resident cells, as well as macrophage infiltration, were significantly suppressed in the lungs of IL-4 KO mice. We also show that IL-4 was significantly increased in the lungs of HIMF-treated WT mice. Our in vitro studies using pulmonary microvascular endothelial cells revealed that HIMF stimulated cell proliferation, vascular endothelial growth factor expression, and MCP-1 production in a manner that is dependent on the IL-4/IL-4Rα system. These findings suggest that IL-4 signaling may play a significant role in HIMF-induced lung inflammation and vascular remodeling.


Respiratory Research | 2013

Hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMα) in chronic hypoxia- and antigen-mediated pulmonary vascular remodeling

Daniel J. Angelini; Qingning Su; Kazuyo Yamaji-Kegan; Chunling Fan; John Skinner; Andre Poloczek; Hazim El-Haddad; Chris Cheadle; Roger A. Johns

BackgroundBoth chronic hypoxia and allergic inflammation induce vascular remodeling in the lung, but only chronic hypoxia appears to cause PH. We investigate the nature of the vascular remodeling and the expression and role of hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMα) in explaining this differential response.MethodsWe induced pulmonary vascular remodeling through either chronic hypoxia or antigen sensitization and challenge. Mice were evaluated for markers of PH and pulmonary vascular remodeling throughout the lung vascular bed as well as HIMF expression and genomic analysis of whole lung.ResultsChronic hypoxia increased both mean pulmonary artery pressure (mPAP) and right ventricular (RV) hypertrophy; these changes were associated with increased muscularization and thickening of small pulmonary vessels throughout the lung vascular bed. Allergic inflammation, by contrast, had minimal effect on mPAP and produced no RV hypertrophy. Only peribronchial vessels were significantly thickened, and vessels within the lung periphery did not become muscularized. Genomic analysis revealed that HIMF was the most consistently upregulated gene in the lungs following both chronic hypoxia and antigen challenge. HIMF was upregulated in the airway epithelial and inflammatory cells in both models, but only chronic hypoxia induced HIMF upregulation in vascular tissue.ConclusionsThe results show that pulmonary vascular remodeling in mice induced by chronic hypoxia or antigen challenge is associated with marked increases in HIMF expression. The lack of HIMF expression in the vasculature of the lung and no vascular remodeling in the peripheral resistance vessels of the lung is likely to account for the failure to develop PH in the allergic inflammation model.


American Journal of Respiratory Cell and Molecular Biology | 2009

Resistin-Like Molecule-β in Scleroderma-Associated Pulmonary Hypertension

Daniel J. Angelini; Qingning Su; Kazuyo Yamaji-Kegan; Chunling Fan; Xingwu Teng; Paul M. Hassoun; Stephen C. Yang; Hunter C. Champion; Rubin M. Tuder; Roger A. Johns

Scleroderma is a systemic, mixed connective tissue disease that can impact the lungs through pulmonary fibrosis, vascular remodeling, and the development of pulmonary hypertension and right heart failure. Currently, little is known about the molecular mechanisms that drive this condition, but we have recently identified a novel gene product that is up-regulated in a murine model of hypoxia-induced pulmonary hypertension. This molecule, known as hypoxia-induced mitogenic factor (HIMF), is a member of the newly described resistin gene family. We have demonstrated that HIMF has mitogenic, angiogenic, vasoconstrictive, inflammatory, and chemokine-like properties, all of which are associated with vascular remodeling in the lung. Here, we demonstrate that the human homolog of HIMF, resistin-like molecule (RELM)-beta, is expressed in the lung tissue of patients with scleroderma-associated pulmonary hypertension and is up-regulated compared with normal control subjects. Immunofluorescence colocalization revealed that RELM-beta is expressed in the endothelium and vascular smooth muscle of remodeled vessels, as well as in plexiform lesions, macrophages, T cells, and myofibroblast-like cells. We also show that addition of recombinant RELM-beta induces proliferation and activation of ERK1/2 in primary cultured human pulmonary endothelial and smooth muscle cells. These results suggest that RELM-beta may be involved in the development of scleroderma-associated pulmonary hypertension.


PLOS ONE | 2010

Hypoxia-Induced Mitogenic Factor (HIMF/FIZZ1/RELMα) Recruits Bone Marrow-Derived Cells to the Murine Pulmonary Vasculature

Daniel J. Angelini; Qingning Su; Irina A. Kolosova; Chunling Fan; John Skinner; Kazuyo Yamaji-Kegan; Michael I. Collector; Saul J. Sharkis; Roger A. Johns

Background Pulmonary hypertension (PH) is a disease of multiple etiologies with several common pathological features, including inflammation and pulmonary vascular remodeling. Recent evidence has suggested a potential role for the recruitment of bone marrow-derived (BMD) progenitor cells to this remodeling process. We recently demonstrated that hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMα) is chemotactic to murine bone marrow cells in vitro and involved in pulmonary vascular remodeling in vivo. Methodology/Principal Findings We used a mouse bone marrow transplant model in which lethally irradiated mice were rescued with bone marrow transplanted from green fluorescent protein (GFP)+ transgenic mice to determine the role of HIMF in recruiting BMD cells to the lung vasculature during PH development. Exposure to chronic hypoxia and pulmonary gene transfer of HIMF were used to induce PH. Both models resulted in markedly increased numbers of BMD cells in and around the pulmonary vasculature; in several neomuscularized small (∼20 µm) capillary-like vessels, an entirely new medial wall was made up of these cells. We found these GFP+ BMD cells to be positive for stem cell antigen-1 and c-kit, but negative for CD31 and CD34. Several of the GFP+ cells that localized to the pulmonary vasculature were α-smooth muscle actin+ and localized to the media layer of the vessels. This finding suggests that these cells are of mesenchymal origin and differentiate toward myofibroblast and vascular smooth muscle. Structural location in the media of small vessels suggests a functional role in the lung vasculature. To examine a potential mechanism for HIMF-dependent recruitment of mesenchymal stem cells to the pulmonary vasculature, we performed a cell migration assay using cultured human mesenchymal stem cells (HMSCs). The addition of recombinant HIMF induced migration of HMSCs in a phosphoinosotide-3-kinase-dependent manner. Conclusions/Significance These results demonstrate HIMF-dependent recruitment of BMD mesenchymal-like cells to the remodeling pulmonary vasculature.


Journal of Immunology | 2009

IL-4 Is Proangiogenic in the Lung under Hypoxic Conditions

Kazuyo Yamaji-Kegan; Qingning Su; Daniel J. Angelini; Roger A. Johns

IL-4-mediated proangiogenic and proinflammatory vascular responses have been implicated in the pathogenesis of chronic lung diseases such as asthma. Although it is well known that hypoxia induces pulmonary angiogenesis and vascular alterations, the underlying mechanism of IL-4 on the pulmonary vasculature under hypoxic conditions remains unknown. In this context, we designed the present study to determine the functional importance of IL-4 for pulmonary angiogenesis under hypoxic conditions using IL-4 knockout (KO) animals. Our results show that hypoxia significantly increased IL-4Rα expression in wild-type (WT) control lungs. Even though hypoxia significantly up-regulated vascular endothelial growth factor (VEGF) receptor expression in the lungs of both genotypes, hypoxia-induced VEGF, VCAM-1, HIF-1α, and ERK phosphorylation were significantly diminished in IL-4 KO lungs as compared with WT control lungs. In addition, hypoxia-induced pulmonary angiogenesis and proliferating activities in the airway and pulmonary artery were significantly suppressed in IL-4 KO lungs as compared with WT control lungs. We also isolated primary lung fibroblasts from these genotypes and stimulated these cells with hypoxia. Hypoxia-induced VEGF production was significantly suppressed in lung fibroblasts from IL-4 KO mice. These in vitro results are in accordance with the in vivo data. Furthermore, we observed a significant increase of hypoxia-induced pulmonary angiogenesis in STAT6 KO mice similar to that in WT controls. In conclusion, IL-4 has proangiogenic properties in the lung under hypoxic conditions via the VEGF pathway, and this is independent of the STAT6 pathway.


American Journal of Respiratory Cell and Molecular Biology | 2015

Resistin-Like Molecule α in Allergen-Induced Pulmonary Vascular Remodeling

Chunling Fan; Lucas W. Meuchel; Qingning Su; Daniel J. Angelini; Ailan Zhang; Chris Cheadle; Irina A. Kolosova; Oleg D. Makarevich; Kazuyo Yamaji-Kegan; Marc E. Rothenberg; Roger A. Johns

Resistin-like molecule α (RELMα) has mitogenic, angiogenic, vasoconstrictive, and chemokine-like properties and is highly relevant in lung pathology. Here, we used RELMα knockout (Retnla(-/-)) mice to investigate the role of RELMα in pulmonary vascular remodeling after intermittent ovalbumin (OVA) challenge. We compared saline- and OVA-exposed wild-type (WT) mice and found that OVA induced significant increases in right ventricular systolic pressure, cardiac hypertrophy, pulmonary vascular remodeling of intra-alveolar arteries, goblet cell hyperplasia in airway epithelium, and intensive lung inflammation, especially perivascular inflammation. Genetic ablation of Retnla prevented the OVA-induced increase in pulmonary pressure and cardiac hypertrophy seen in WT mice. Histological analysis showed that Retnla(-/-) mice exhibited less vessel muscularization, less perivascular inflammation, reduced medial thickness of intra-alveolar vessels, and fewer goblet cells in upper airway epithelium (250-600 μm) than did WT animals after OVA challenge. Gene expression profiles showed that genes associated with vascular remodeling, including those related to muscle protein, contractile fibers, and actin cytoskeleton, were expressed at a lower level in OVA-challenged Retnla(-/-) mice than in similarly treated WT mice. In addition, bronchoalveolar lavage from OVA-challenged Retnla(-/-) mice had lower levels of cytokines, such as IL-1β, -1 receptor antagonist, and -16, chemokine (C-X-C motif) ligand 1, -2, -9, -10, and -13, monocyte chemoattractant protein-1, macrophage colony-stimulating factor, TIMP metallopeptidase inhibitor-1, and triggering receptor expressed on myeloid cells-1, than did that from WT mice when analyzed by cytokine array dot blots. Retnla knockout inhibited the OVA-induced T helper 17 response but not the T helper 2 response. Altogether, our results suggest that RELMα is involved in immune response-induced pulmonary vascular remodeling and the associated increase in inflammation typically observed after OVA challenge.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2009

Unveiling cell phenotypes in lung vascular remodeling

Roger A. Johns; Kazuyo Yamaji-Kegan

PULMONARY VASCULAR REMODELING is a hallmark of most forms of pulmonary hypertension (PH), both primary and secondary. This remodeling takes the form of concentric medial thickening of small arterioles, neomuscularization of previously nonmuscular capillary-like vessels, and structural wall changes in larger pulmonary arteries. The pulmonary arterial muscularization is characterized by increased numbers of vascular smooth muscle cells (SMCs) as well as by hypertrophy of individual SMCs (25). In human PH, characteristic plexiform lesions develop, and there is increasing discussion of neovasculogenesis and angiogenesis during remodeling. The vascular remodeling is associated with hypoxia- or inflammationinduced production of growth factors, angiogenic factors, inflammatory mediators, and vasoconstrictors. For decades, investigators have attempted to dissect the mechanisms behind this lung-specific vascular remodeling, investigating a wide range of environmental stimuli, hemodynamic events, biochemical and molecular signaling pathways, and extracellular matrix changes. Recently, investigators have begun looking in greater detail at the heterogeneity of cells in lung vascular walls under normal and pathological remodeling conditions (14, 26). Initially, it was recognized that even within a single region, endothelial cells differ and may have highly distinct characteristics and functions. Now there is increasing interest in and evidence for the existence of diverse cell types within the media of remodeling lung vessels. Emerging evidence suggests that endogenous or circulating inflammatory and/or progenitor cells contribute significantly to the remodeling process (9, 22). The term “circulating inflammatory/progenitor cells” characterizes a wide variety of cell populations that differ with respect to their structural characteristics, expression of marker molecules, and biological functions. These cells include macrophages and other inflammatory cells, mononuclear cells, stem cells, endothelial progenitor cells, fibrocytes, and myofibroblasts. The nature of these cells, their relative importance, and their derivation and source continue to be unclear and highly controversial. The phenotype of these cells may change during the remodeling process or vary with the specific initiating pathophysiology or type of PH. Thus, it is important to define the phenotype of cells in the vascular media and the roles of the cells in the normal and diseased pulmonary vasculature. The exact source of the cells is also important to know because it may determine cell phenotype and targets for


American Journal of Physiology-lung Cellular and Molecular Physiology | 2006

Hypoxia-induced mitogenic factor has proangiogenic and proinflammatory effects in the lung via VEGF and VEGF receptor-2

Kazuyo Yamaji-Kegan; Qingning Su; Daniel J. Angelini; Hunter C. Champion; Roger A. Johns


american thoracic society international conference | 2009

Hypoxia-Induced Mitogenic Factor (HIMF) Is Involved in the Pulmonary Vascular Remodeling Associated with Chronic Hypoxia and Inflammatory Asthma.

Daniel J. Angelini; Qingning Su; Kazuyo Yamaji-Kegan; Chunling Fan; John Skinner; H El-Haddad; Hc Champion; Roger A. Johns

Collaboration


Dive into the Kazuyo Yamaji-Kegan's collaboration.

Top Co-Authors

Avatar

Roger A. Johns

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chunling Fan

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Chris Cheadle

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

John Skinner

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qingning Su

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Qingning Su

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Allen C. Myers

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge