Network


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

Hotspot


Dive into the research topics where Janusz H. Kabarowski is active.

Publication


Featured researches published by Janusz H. Kabarowski.


Prostaglandins & Other Lipid Mediators | 2009

G2A and LPC: Regulatory functions in immunity

Janusz H. Kabarowski

The G2A receptor was originally identified by virtue of its transcriptional induction in murine B lymphoid cells in response to oncogenic transformation and treatment with various DNA-damaging agents. While preliminary characterization of cellular responses to G2A overexpression in fibroblastic cell lines suggested that this receptor may negatively regulate cell growth under conditions of proliferative and genotoxic stress, subsequent studies driven by the discovery of lysophosphatidylcholine (LPC) as a regulator of G2A signaling in immunoregulatory cells point to an important role for this receptor in innate and adaptive immunity.


American Journal of Physiology-heart and Circulatory Physiology | 2015

Obesity superimposed on aging magnifies inflammation and delays the resolving response after myocardial infarction

Elizabeth Lopez; Janusz H. Kabarowski; Kevin A. Ingle; Vasundhara Kain; Stephen Barnes; David K. Crossman; Merry L. Lindsey; Ganesh V. Halade

Polyunsaturated fatty acid (PUFA) intake has increased over the last 100 yr, contributing to the current obesogenic environment. Obesity and aging are prominent risk factors for myocardial infarction (MI). How obesity interacts with aging to alter the post-MI response, however, is unclear. We tested the hypothesis that obesity in aging mice would impair the resolution of post-MI inflammation. PUFA diet (PUFA aging group) feeding to 12-mo-old C57BL/6J mice for 5 mo showed higher fat mass compared with standard lab chow (LC)-fed young (LC young group; 3-5 mo old) or aging alone control mice (LC aging group). LC young, LC aging, and PUFA aging mice were subjected to coronary artery ligation to induce MI. Despite similar infarct areas post-MI, plasma proteomic profiling revealed higher VCAM-1 in the PUFA aging group compared with LC young and LC aging groups, leading to increased neutrophil infiltration in the PUFA aging group (P<0.05). Macrophage inflammatory protein-1γ and CD40 were also increased at day 1, and myeloperoxidase remained elevated at day 5, an observation consistent with delayed wound healing in the PUFA aging group. Lipidomic analysis showed higher levels of arachidonic acid and 12(S)-hydroxyeicosatetraenoic acid at day 1 post-MI in the PUFA aging group compared with the LC aging group (all P<0.05), thereby mediating neutrophil extravasation in the PUFA aging group. The inflammation-resolving enzymes 5-lipoxygenase, cyclooxygenase-2, and heme oxyegnase-1 were altered to delay wound healing post-MI in the PUFA aging group compared with LC young and LC aging groups. PUFA aging magnifies the post-MI inflammatory response and impairs the healing response by stimulating prolonged neutrophil trafficking and proinflammatory lipid mediators.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2006

Loss of the Lysophosphatidylcholine Effector, G2A, Ameliorates Aortic Atherosclerosis in Low-Density Lipoprotein Receptor Knockout Mice

Brian W. Parks; Aldons J. Lusis; Janusz H. Kabarowski

Objective—Lysophosphatidylcholine is a major product of low-density lipoprotein (LDL) oxidation and secretory phospholipase A2-mediated lipid hydrolysis within atherosclerotic lesions. The G2A receptor mediates chemotaxis of cultured macrophages and T cells to lysophosphatidylcholine, supporting a pro-atherogenic role for this receptor in vivo. We investigated the ability of G2A to modulate atherosclerosis in mice. Methods and Results—We measured atherosclerosis in G2A+/+ and G2A−/− LDL receptor knockout (LDLR−/−) mice. Consistent with a previous study, early lesion size at the aortic sinus was unaffected by G2A deficiency. However, G2A deficiency attenuated lesion progression at this site (42% to 44% reduction in average lesion area) and led to robust suppression of atherosclerosis throughout the aorta after short and extended periods of diet intervention (reduction in aortic lesion coverage: 62% to 73% at 9 weeks, 75% to 84% at 20 weeks). In G2A−/−LDLR−/− mice, intimal macrophage accumulation at lesion-prone sites of the aorta was significantly reduced in the absence of any detectable effect on T cell recruitment. Examination of lipoprotein profiles revealed elevated levels of circulating high-density lipoprotein (HDL) cholesterol in G2A−/−LDLR−/− mice compared with their G2A+/+LDLR−/− counterparts after extended periods of diet intervention (54% increase in mean HDL cholesterol concentration). Conclusion—G2A provides a pro-atherogenic stimulus in vivo consistent with its chemotactic action but to which a pleiotropy of effects, including modulation of lipoprotein metabolism, may also contribute.


Arthritis & Rheumatism | 2011

Autoimmune-mediated reduction of high-density lipoprotein–cholesterol and paraoxonase 1 activity in systemic lupus erythematosus–prone gld mice

Roshni Srivastava; Shaohua Yu; Brian W. Parks; Leland L. Black; Janusz H. Kabarowski

OBJECTIVE To characterize modifications of high-density lipoprotein (HDL) in autoimmune gld mice that may be relevant to premature atherosclerosis in systemic lupus erythematosus, and to assess their relationship to specific aspects of autoimmune disease. METHODS HDL cholesterol (HDL-C), apolipoprotein A-I (Apo A-I), paraoxonase 1 (PON1) activity, hepatic gene expression, and HDL biogenesis were measured in aging female gld and wild-type congenic mice. Autoantibodies, lymphoid organs, and cytokines were analyzed by enzyme-linked immunosorbent assay, flow cytometry, and multiplex assay, respectively. RESULTS Plasma HDL-C, HDL Apo A-I, and HDL-associated PON1 activity were reduced in aging gld mice in association with the development of autoimmunity, independent of changes in hepatic Apo A-I and PON1 expression or HDL biogenesis. Hepatic induction of the acute-phase reactant serum amyloid A1 resulted in its incorporation into HDL in gld mice. Deletion of the lipid-sensitive receptor G2A in gld mice (G2A-/- gld) attenuated reductions in HDL-C and PON1 activity without altering hepatic Apo A-I and PON1 expression, HDL biogenesis, or levels of acute-phase proinflammatory cytokines. Plasma anti-Apo A-I autoantibodies were elevated in aging gld mice commensurate with detectable increases in Apo A-I immune complexes. Autoantibody levels were lower in aging G2A-/- gld mice compared with gld mice, and anti-Apo A-I autoantibody levels were significantly related to HDL-C concentrations (r=-0.645, P<0.00004) and PON1 activity (r=-0.555, P<0.0007) among autoimmune gld and G2A-/- gld mice. CONCLUSION Autoantibodies against Apo A-I contribute to reducing HDL-C and PON1 activity in autoimmune gld mice independently of hepatic HDL biogenesis, suggesting that functional impairment and premature clearance of HDL immune complexes may be principal mechanisms involved.


Journal of Clinical Investigation | 2015

Interferon-induced mechanosensing defects impede apoptotic cell clearance in lupus.

Hao Li; Yang-Xin Fu; Qi Wu; Yong Zhou; David K. Crossman; PingAr Yang; Jun Li; Bao Luo; Laurence Morel; Janusz H. Kabarowski; Hideo Yagita; Carl F. Ware; Hui-Chen Hsu; John D. Mountz

Systemic lupus erythematosus (SLE) is a severe autoimmune disease that is associated with increased circulating apoptotic cell autoantigens (AC-Ags) as well as increased type I IFN signaling. Here, we describe a pathogenic mechanism in which follicular translocation of marginal zone (MZ) B cells in the spleens of BXD2 lupus mice disrupts marginal zone macrophages (MZMs), which normally clear AC debris and prevent follicular entry of AC-Ags. Phagocytosis of ACs by splenic MZMs required the megakaryoblastic leukemia 1 (MKL1) transcriptional coactivator-mediated mechanosensing pathway, which was maintained by MZ B cells through expression of membrane lymphotoxin-α1β2 (mLT). Specifically, type I IFN-induced follicular shuttling of mLT-expressing MZ B cells disengaged interactions between these MZ B cells and LTβ receptor-expressing MZMs, thereby downregulating MKL1 in MZMs. Loss of MKL1 expression in MZMs led to defective F-actin polymerization, inability to clear ACs, and, eventually, MZM dissipation. Aggregation of plasmacytoid DCs in the splenic perifollicular region, follicular translocation of MZ B cells, and loss of MKL1 and MZMs were also observed in an additional murine lupus model and in the spleens of patients with SLE. Collectively, the results suggest that lupus might be interrupted by strategies that maintain or enhance mechanosensing signaling in the MZM barrier to prevent follicular entry of AC-Ags.


Radiation Research | 2011

Iron-Ion Radiation Accelerates Atherosclerosis in Apolipoprotein E-Deficient Mice

Tao Yu; Brian W. Parks; Shaohua Yu; Roshni Srivastava; Kiran Gupta; Xing Wu; Saman Khaled; Polly Y. Chang; Janusz H. Kabarowski; Dennis F. Kucik

Abstract Radiation exposure from a number of terrestrial sources is associated with an increased risk for atherosclerosis. Recently, concern over whether exposure to cosmic radiation might pose a similar risk for astronauts has increased. To address this question, we examined the effect of 2 to 5 Gy iron ions (56Fe), a particularly damaging component of cosmic radiation, targeted to specific arterial sites in male apolipoprotein E-deficient (apoE−/−) mice. Radiation accelerated the development of atherosclerosis in irradiated portions of the aorta independent of any systemic effects on plasma lipid profiles or circulating leukocytes. Further, radiation exposure resulted in a more rapid progression of advanced aortic root lesions, characterized by larger necrotic cores associated with greater numbers of apoptotic macrophages and reduced lesional collagen compared to sham-treated mice. Intima media thickening of the carotid arteries was also exacerbated. Exposure to 56Fe ions can therefore accelerate the development of atherosclerotic lesions and promote their progression to an advanced stage characterized by compositional changes indicative of increased thrombogenicity and instability. We conclude that the potential consequences of radiation exposure for astronauts on prolonged deep-space missions are a major concern. Knowledge gained from further studies with animal models should lead to a better understanding of the pathophysiological effects of accelerated ion radiation to better estimate atherogenic risk and develop appropriate countermeasures to mitigate its damaging effects.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2009

ApoE-Dependent Modulation of HDL and Atherosclerosis by G2A in LDL Receptor–Deficient Mice Independent of Bone Marrow–Derived Cells

Brian W. Parks; Roshni Srivastava; Shaohua Yu; Janusz H. Kabarowski

Objective—Deletion of the lysophospholipid-sensitive receptor, G2A, in low-density lipoprotein receptor knockout (LDLR−/−) mice elevates plasma high-density lipoprotein (HDL) cholesterol and suppresses atherosclerosis. However, chemotactic action of G2A in monocytes/macrophages, in addition to its modulatory effect on HDL, may contribute to the proatherogenic action of G2A. Methods and Results—We determined that deletion of G2A in LDLR−/− mice increases the ApoA1, ApoE, and cholesterol content of plasma HDL fractions. Hepatocytes were shown to express G2A and hepatocytes from G2A-deficient LDLR−/− mice secreted more ApoA1 and ApoE in HDL fractions compared to their G2A-sufficient counterparts. The atheroprotective and HDL modulatory effects of G2A deficiency were dependent on the presence of ApoE, as deletion of G2A in ApoE−/− and ApoE−/−LDLR−/− mice failed to raise HDL and did not suppress atherosclerosis. G2A deficiency in bone marrow–derived cells of LDLR−/− mice had no effect on atherosclerosis or HDL, whereas G2A deficiency in resident tissues was sufficient to raise HDL and suppress atherosclerosis. Conclusion—These data demonstrate that the chemotactic function of G2A in bone marrow–derived monocytes does not modulate atherosclerosis in LDLR−/− mice and suggest an ApoE-dependent function for G2A in the control of hepatic HDL metabolism that might contribute to its proatherogenic action.


American Journal of Physiology-renal Physiology | 2016

Early lipid changes in acute kidney injury using SWATH lipidomics coupled with MALDI tissue imaging.

Sangeetha Rao; Kelly Walters; Landon Wilson; Bo Chen; Subhashini Bolisetty; David E. Graves; Stephen Barnes; Anupam Agarwal; Janusz H. Kabarowski

Acute kidney injury (AKI) is one of the leading causes of in-hospital morbidity and mortality, particularly in critically ill patients. Although our understanding of AKI at the molecular level remains limited due to its complex pathophysiology, recent advances in both quantitative and spatial mass spectrometric approaches offer new opportunities to assess the significance of renal metabolomic changes in AKI models. In this study, we evaluated lipid changes in early ischemia-reperfusion (IR)-related AKI in mice by using sequential window acquisition of all theoretical spectra (SWATH)-mass spectrometry (MS) lipidomics. We found a significant increase in two abundant ether-linked phospholipids following IR at 6 h postinjury, a plasmanyl choline, phosphatidylcholine (PC) O-38:1 (O-18:0, 20:1), and a plasmalogen, phosphatidylethanolamine (PE) O-42:3 (O-20:1, 22:2). Both of these lipids correlated with the severity of AKI as measured by plasma creatinine. In addition to many more renal lipid changes associated with more severe AKI, PC O-38:1 elevations were maintained at 24 h post-IR, while renal PE O-42:3 levels decreased, as were all ether PEs detected by SWATH-MS at this later time point. To further assess the significance of this early increase in PC O-38:1, we used matrix-assisted laser desorption ionization imaging mass spectrometry (MALDI-IMS) to determine that it occurred in proximal tubules, a region of the kidney that is most prone to IR injury and also rich in the rate-limiting enzymes involved in ether-linked phospholipid biosynthesis. Use of SWATH-MS lipidomics in conjunction with MALDI-IMS for lipid localization will help in elucidating the role of lipids in the pathobiology of AKI.


Journal of Lipid Research | 2013

CD36, but not G2A, modulates efferocytosis, inflammation and fibrosis following bleomycin-induced lung injury

Brian W. Parks; Leland L. Black; Kurt A. Zimmerman; Allison E. Metz; Chad Steele; Joanne E. Murphy-Ullrich; Janusz H. Kabarowski

Macrophage G2A and CD36 lipid receptors are thought to mediate efferocytosis following tissue injury and thereby prevent excessive inflammation that could compromise tissue repair. To test this, we subjected mice lacking G2A or CD36 receptor to bleomycin-induced lung injury and measured efferocytosis, inflammation, and fibrosis. Loss of CD36 (but not G2A) delayed clearance of apoptotic alveolar cells (mean 78% increase in apoptotic cells 7 days postinjury), potentiated inflammation (mean 56% increase in lung neutrophils and 75% increase in lung KC levels 7 days postinjury, 51% increase in lung macrophages 14 days postinjury), and reduced lung fibrosis (mean 41% and 29% reduction 14 and 21 days postinjury, respectively). Reduced fibrosis in CD36−/− mice was associated with lower levels of profibrotic TH2 cytokines (IL-9, IL-13, IL-4), decreased expression of the M2 macrophage marker Arginase-1, and reduced interstitial myofibroblasts. G2A, on the other hand, was required for optimal clearance of apoptotic neutrophils during zymosan-induced peritoneal inflammation (50.3% increase in apoptotic neutrophils and 30.6% increase in total neutrophils 24 h following zymosan administration in G2A−/− mice). Thus, CD36 is required for timely removal of apoptotic cells in the context of lung injury and modulates subsequent inflammatory and fibrotic processes relevant to fibrotic lung disease.


Journal of Mass Spectrometry | 2016

Training in metabolomics research. I. Designing the experiment, collecting and extracting samples and generating metabolomics data: Design and execution of a metabolomics experiment

Stephen Barnes; H. Paul Benton; Krista Casazza; Sara J. Cooper; Xiangqin Cui; Xiuxia Du; Jeffrey A. Engler; Janusz H. Kabarowski; Shuzhao Li; Wimal Pathmasiri; Jeevan K. Prasain; Matthew B. Renfrow; Hemant K. Tiwari

The study of metabolism has had a long history. Metabolomics, a systems biology discipline representing analysis of known and unknown pathways of metabolism, has grown tremendously over the past 20 years. Because of its comprehensive nature, metabolomics requires careful consideration of the question(s) being asked, the scale needed to answer the question(s), collection and storage of the sample specimens, methods for extraction of the metabolites from biological matrices, the analytical method(s) to be employed and the quality control of the analyses, how collected data are correlated, the statistical methods to determine metabolites undergoing significant change, putative identification of metabolites and the use of stable isotopes to aid in verifying metabolite identity and establishing pathway connections and fluxes. The National Institutes of Health Common Fund Metabolomics Program was established in 2012 to stimulate interest in the approaches and technologies of metabolomics. To deliver one of the programs goals, the University of Alabama at Birmingham has hosted an annual 4-day short course in metabolomics for faculty, postdoctoral fellows and graduate students from national and international institutions. This paper is the first part of a summary of the training materials presented in the course to be used as a resource for all those embarking on metabolomics research. The complete set of training materials including slide sets and videos can be viewed at http://www.uab.edu/proteomics/metabolomics/workshop/workshop_june_2015.php. Copyright

Collaboration


Dive into the Janusz H. Kabarowski's collaboration.

Top Co-Authors

Avatar

Stephen Barnes

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Brian W. Parks

University of California

View shared research outputs
Top Co-Authors

Avatar

Shaohua Yu

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Dennis F. Kucik

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Kiran Gupta

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Leland L. Black

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Roshni Srivastava

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Tao Yu

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Ganesh V. Halade

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Xing Wu

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge