Network


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

Hotspot


Dive into the research topics where Michael Miksa is active.

Publication


Featured researches published by Michael Miksa.


Journal of Immunology | 2009

Milk Fat Globule Epidermal Growth Factor-Factor VIII Is Down-Regulated in Sepsis via the Lipopolysaccharide-CD14 Pathway

Hidefumi Komura; Michael Miksa; Rongqian Wu; Sanna M. Goyert; Ping Wang

Phagocytosis prevents the release of potentially harmful or immunogenic materials from dying cells. Milk fat globule epidermal growth factor (EGF)-factor VIII (MFG-E8) mediates the clearance of apoptotic cells. We have previously shown that the administration of MFG-E8-rich exosomes from immature dendritic cells promotes the phagocytosis of apoptotic cells and improves survival in sepsis. Because endotoxin is elevated in polymicrobial sepsis, we hypothesized that down-regulation of MFG-E8 is mediated via the LPS-CD14 pathway, eventually leading to the accruement of apoptotic cells. Polymicrobial sepsis was induced by cecal ligation and puncture (CLP) in CD14-deficient (CD14−/−), TLR4-mutated and wild-type (WT) mice. In addition, endotoxemia was elicited by i.p. injection of LPS. LPS was also neutralized by pretreating CLP-induced WT mice with polymyxin B. Splenic MFG-E8 expression, phagocytic activity, and apoptosis were assessed 5 and 20 h after CLP or 5 h after LPS administration. In septic WT mice, MFG-E8 mRNA and protein levels were suppressed by 49 and 33%, respectively. Endotoxemia reduced MFG-E8 mRNA expression in a dose dependent manner and the down-regulation of MFG-E8 mRNA expression in CLP-induced sepsis was attenuated by polymyxin B. This CLP-induced suppression was not observed in both CD14−/− and TLR4-mutated mice. CLP significantly decreased phagocytic activity of peritoneal macrophages in WT (by 30%), but not in CD14−/− mice. CLP also induced significant apoptosis in the spleen of WT (by 61%), but less in CD14−/− mice. Thus, MFG-E8 production is down-regulated in sepsis by LPS-CD14 dependent fashion, leading to a reduction of phagocytosis of apoptotic cells.


PLOS ONE | 2009

Pivotal Role of the α2A-Adrenoceptor in Producing Inflammation and Organ Injury in a Rat Model of Sepsis

Michael Miksa; Padmalaya Das; Mian Zhou; Rongqian Wu; Weifeng Dong; Youxin Ji; Sanna M. Goyert; Thanjavur S. Ravikumar; Ping Wang

Background Norepinephrine (NE) modulates the responsiveness of macrophages to proinflammatory stimuli through the activation of adrenergic receptors (ARs). Being part of the stress response, early increases of NE in sepsis sustain adverse systemic inflammatory responses. The intestine is an important source of NE release in the early stage of cecal ligation and puncture (CLP)-induced sepsis in rats, which then stimulates TNF-α production in Kupffer cells (KCs) through the activation of the α2-AR. It is important to know which of the three α2-AR subtypes (i.e., α2A, α2B or α2C) is responsible for the upregulation of TNF-α production. The aim of this study was to determine the contribution of α2A-AR in this process. Methodology/Principal Findings Adult male rats underwent CLP and KCs were isolated 2 h later. Gene expression of α2A-AR was determined. In additional experiments, cultured KCs were incubated with NE with or without BRL-44408 maleate, a specific α2A-AR antagonist, and intraportal infusion of NE for 2 h with or without BRL-44408 maleate was carried out in normal animals. Finally, the impact of α2A-AR activation by NE was investigated under inflammatory conditions (i.e., endotoxemia and CLP). Gene expression of the α2A-AR subtype was significantly upregulated after CLP. NE increased the release of TNF-α in cultured KCs, which was specifically inhibited by the α2A-AR antagonist BRL-44408. Equally, intraportal NE infusion increased TNF-α gene expression in KCs and plasma TNF-α which was also abrogated by co-administration of BRL-44408. NE also potentiated LPS-induced TNF-α release via the α2A-AR in vitro and in vivo. This potentiation of TNF-α release by NE was mediated through the α2A-AR coupled Gαi protein and the activation of the p38 MAP kinase. Treatment of septic animals with BRL-44408 suppressed TNF-α, prevented multiple organ injury and significantly improved survival from 45% to 75%. Conclusions/Significance Our novel finding is that hyperresponsiveness to α2-AR stimulation observed in sepsis is primarily due to an increase in α2A-AR expression in KCs. This appears to be in part responsible for the increased proinflammatory response and ensuing organ injury in sepsis. These findings provide important feasibility information for further developing the α2A-AR antagonist as a new therapy for sepsis.


Critical Care | 2009

VX-166: a novel potent small molecule caspase inhibitor as a potential therapy for sepsis

Peter Weber; Ping Wang; Stephane Maddens; Paul Wang; Rongqian Wu; Michael Miksa; Weifeng Dong; Michael Mortimore; Julian Golec; Peter Charlton

IntroductionPrevention of lymphocyte apoptosis by caspase inhibition has been proposed as a novel treatment approach in sepsis. However, it has not been clearly demonstrated that caspase inhibitors improve survival in sepsis models when dosed post-insult. Also, there are concerns that caspase inhibitors might suppress the immune response. Here we characterize VX-166, a broad caspase inhibitor, as a novel potential treatment for sepsis.MethodsVX-166 was studied in a number of enzymatic and cellular assays. The compound was then tested in a murine model of endotoxic shock (lipopolysaccharide (LPS), 20 mg/kg IV) and a 10 d rat model of polymicrobial sepsis by caecal ligation and puncture (CLP).ResultsVX-166 showed potent anti-apoptotic activity in vitro and inhibited the release of interleukin (IL)-1beta and IL-18. In the LPS model, VX-166 administered 0, 4, 8 and 12 h post-LPS significantly improved survival in a dose-dependent fashion (P < 0.0028). In the CLP model, VX-166 continuously administered by mini-osmotic pump significantly improved survival when dosed 3 h after insult, (40% to 92%, P = 0.009). When dosed 8 h post-CLP, VX-166 improved survival from 40% to 66% (P = 0.19). Mode of action studies in the CLP model confirmed that VX-166 significantly inhibited thymic atrophy and lymphocyte apoptosis as determined by flow cytometry (P < 0.01). VX-166 reduced plasma endotoxin levels (P < 0.05), suggesting an improved clearance of bacteria from the bloodstream. Release of IL-1beta in vivo or T-cell activation in vitro were moderately affected.ConclusionsOur studies enhance the case for the use of caspase inhibitors in sepsis. VX-166 itself has promise as a therapy for the treatment of sepsis in man.


Critical Care Medicine | 2015

1224: AN ATYPICAL PRESENTATION OF OTC DEFICIENCY IN AN ADOLESCENT MALE

Mark Shlomovich; Michael Miksa; Jacqueline Weingarten-Arams; Jennifer Liedel

Crit Care Med 2015 • Volume 43 • Number 12 (Suppl.) She met 6 of 8 criteria for HLH (fever, splenomegaly, cytopenia, hemophagocytosis in bone marrow, elevated serum ferritin and soluble interleukin-2 receptor levels) and was treated with high dose steroids and etoposide. She made gradual recovery and was discharged home 3 weeks later. An uncontrolled hypercytokinemia complicates differentiation of HLH from severe sepsis. However, early investigations can facilitate diagnosis and aggressive treatment of HLH with good outcome.


Journal of Immunology | 2009

Immature Dendritic Cell-Derived Exosomes Rescue Septic Animals via MFGE8

Michael Miksa; Rongqian Wu; Weifeng Dong; Hidefumi Komura; Dhruv Amin; Youxin Ji; Zhimin Wang; Haichao Wang; Thanjavur S. Ravikumar; Kevin J. Tracey; Ping Wang

Sepsis, a highly lethal systemic inflammatory syndrome, is associated with increases of proinflammatory cytokines (e.g., TNF-α, HMGB1) and the accumulation of apoptotic cells that have the potential to be detrimental. Depending on the timing and tissue, prevention of apoptosis in sepsis is beneficial; however thwarting the development of secondary necrosis through the active removal of apoptotic cells by phagocytosis may offer a novel anti-sepsis therapy. Immature dendritic cells (IDCs) release exosomes that contain milk fat globule EGF factor 8 (MFGE8), a protein required to opsonize apoptotic cells for phagocytosis. In an experimental sepsis model using cecal ligation and puncture, we found that MFGE8 levels decreased in the spleen and blood, which was associated with impaired apoptotic cell clearance. Administration of IDC-derived exosomes promoted phagocytosis of apoptotic cells and significantly reduced mortality. Treatment with recombinant MFGE8 was equally protective, while MFGE8-deficient mice suffered from increased mortality. IDC exosomes also attenuated the release of proinflammatory cytokines in septic rats. Liberation of HMGB1, a nuclear protein that contributes to inflammation upon release from unengulfed apoptotic cells, was prevented by MFGE8-mediated phagocytosis in vitro. We conclude that IDC-derived exosomes attenuate the acute systemic inflammatory response in sepsis by enhancing apoptotic cell clearance via MFGE8.Sepsis, a highly lethal systemic inflammatory syndrome, is associated with increases of proinflammatory cytokines (e.g., TNF-α, HMGB1) and the accumulation of apoptotic cells that have the potential to be detrimental. Depending on the timing and tissue, prevention of apoptosis in sepsis is beneficial; however, thwarting the development of secondary necrosis through the active removal of apoptotic cells by phagocytosis may offer a novel anti-sepsis therapy. Immature dendritic cells (IDCs) release exosomes that contain milk fat globule EGF factor VIII (MFGE8), a protein required to opsonize apoptotic cells for phagocytosis. In an experimental sepsis model using cecal ligation and puncture, we found that MFGE8 levels decreased in the spleen and blood, which was associated with impaired apoptotic cell clearance. Administration of IDC-derived exosomes promoted phagocytosis of apoptotic cells and significantly reduced mortality. Treatment with recombinant MFGE8 was equally protective, whereas MFGE8-deficient mice suffered from increased mortality. IDC exosomes also attenuated the release of proinflammatory cytokines in septic rats. Liberation of HMGB1, a nuclear protein that contributes to inflammation upon release from unengulfed apoptotic cells, was prevented by MFGE8-mediated phagocytosis in vitro. We conclude that IDC-derived exosomes attenuate the acute systemic inflammatory response in sepsis by enhancing apoptotic cell clearance via MFGE8.


Shock | 2006

GHRELIN INHIBITS SYMPATHETIC NERVOUS ACTIVITY IN SEPSIS

Rongqian Wu; Mian Zhou; Padmalaya Das; Weifeng Dong; Y. Ji; Derek Yang; Michael Miksa; T.S. Ravikumar; P. Wang

Our previous studies have shown that norepinephrine (NE) upregulates proinflammatory cytokines by activating alpha(2)-adrenoceptor. Therefore, modulation of the sympathetic nervous system represents a novel treatment for sepsis. We have also shown that a novel stomach-derived peptide, ghrelin, is downregulated in sepsis and that its intravenous administration decreases proinflammatory cytokines and mitigates organ injury. However, it remains unknown whether ghrelin inhibits sympathetic activity through central ghrelin receptors [i.e., growth hormone secretagogue receptor 1a (GHSR-la)] in sepsis. To study this, sepsis was induced in male rats by cecal ligation and puncture (CLP). Ghrelin was administered through intravenous or intracerebroventricular injection 30 min before CLP. Our results showed that intravenous administration of ghrelin significantly reduced the elevated NE and TNF-alpha levels at 2 h after CLP. NE administration partially blocked the inhibitory effect of ghrelin on TNF-alpha in sepsis. GHSR-la inhibition by the administration of a GHSR-la antagonist, [d-Arg(1),d-Phe(5), d-Trp(7,9),Leu(11)]substance P, significantly increased both NE and TNF-alpha levels even in normal animals. Markedly elevated circulating levels of NE 2 h after CLP were also significantly decreased by intracerebroventricular administration of ghrelin. Ghrelins inhibitory effect on NE release was completely blocked by intracerebroventricular injection of the GHSR-1a antagonist or a neuropeptide Y (NPY)/Y(1) receptor antagonist. However, ghrelins downregulatory effect on TNF-alpha release was only partially diminished by these agents. Thus ghrelin has sympathoinhibitory properties that are mediated by central ghrelin receptors involving a NPY/Y1 receptor-dependent pathway. Ghrelins inhibitory effect on TNF-alpha production in sepsis is partially because of its modulation of the overstimulated sympathetic nerve activation.


Shock | 2006

Dendritic cell-derived exosomes containing milk fat globule epidermal growth factor-factor VIII attenuate proinflammatory responses in sepsis

Michael Miksa; Rongqian Wu; Weifeng Dong; Padmalaya Das; Derek Yang; P. Wang


International Journal of Molecular Medicine | 1998

Maturation-induced down-regulation of MFG-E8 impairs apoptotic cell clearance and enhances endotoxin response.

Michael Miksa; Dhruv Amin; Rongqian Wu; Asha Jacob; Mian Zhou; Weifeng Dong; Weng Lang Yang; Thanjavur S. Ravikumar; P. Wang


Shock | 2006

FRACTALKINE INCREASES APOPTOTIC CELL CLEARANCE BY UPREGULATING MFG-E8 IN MACROPHAGES

Michael Miksa; D. Amin; Rongqian Wu; T.S. Ravikumar; P. Wang


Critical Care Medicine | 2018

1317: NONINVASIVE VENTILATION FACTORS ASSOCIATED WITH THE SAFETY OF TRACHEAL INTUBATION

Natalie Napolitano; Guillaume Emeriaud; Lee A. Polikoff; John S. Giuliano; Michael Miksa; Simon Li; Vinay Nadkarni; Akira Nishisaki

Collaboration


Dive into the Michael Miksa's collaboration.

Top Co-Authors

Avatar

Rongqian Wu

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Weifeng Dong

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Ping Wang

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

P. Wang

North Shore-LIJ Health System

View shared research outputs
Top Co-Authors

Avatar

Mian Zhou

Rhode Island Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Youxin Ji

North Shore-LIJ Health System

View shared research outputs
Top Co-Authors

Avatar

Hidefumi Komura

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Simon Li

New York Medical College

View shared research outputs
Top Co-Authors

Avatar

Akira Nishisaki

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge