Network


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

Hotspot


Dive into the research topics where Qi Mi is active.

Publication


Featured researches published by Qi Mi.


Wiley Interdisciplinary Reviews: Systems Biology and Medicine | 2009

Agent-based models in translational systems biology.

Gary An; Qi Mi; Joyeeta Dutta-Moscato; Yoram Vodovotz

Effective translational methodologies for knowledge representation are needed in order to make strides against the constellation of diseases that affect the world today. These diseases are defined by their mechanistic complexity, redundancy, and nonlinearity. Translational systems biology aims to harness the power of computational simulation to streamline drug/device design, simulate clinical trials, and eventually to predict the effects of drugs on individuals. The ability of agent‐based modeling to encompass multiple scales of biological process as well as spatial considerations, coupled with an intuitive modeling paradigm, suggests that this modeling framework is well suited for translational systems biology. This review describes agent‐based modeling and gives examples of its translational applications in the context of acute inflammation and wound healing. Copyright


Wound Repair and Regeneration | 2007

Agent-based model of inflammation and wound healing : insights into diabetic foot ulcer pathology and the role of transforming growth factor-β1

Qi Mi; Béatrice Rivière; Gilles Clermont; David L. Steed; Yoram Vodovotz

Inflammation and wound healing are inextricably linked and complex processes, and are deranged in the setting of chronic, nonhealing diabetic foot ulcers (DFU). An ideal therapy for DFU should both suppress excessive inflammation while enhancing healing. We reasoned that biological simulation would clarify mechanisms and help refine therapeutic approaches to DFU. We developed an agent‐based model (ABM) capable of reproducing qualitatively much of the literature data on skin wound healing, including changes in relevant cell populations (macrophages, neutrophils, fibroblasts) and their key effector cytokines (tumor necrosis factor‐α [TNF], interleukin [IL]‐1β, IL‐10, and transforming growth factor [TGF]‐β1). In this simulation, a normal healing response results in tissue damage that first increases (due to wound‐induced inflammation) and then decreases as the collagen levels increase. Studies by others suggest that diabetes and DFU are characterized by elevated TNF and reduced TGF‐β1, although which of these changes is a cause and which one is an effect is unclear. Accordingly, we simulated the genesis of DFU in two ways, either by (1) increasing the rate of TNF production fourfold or (2) by decreasing the rate of TGF‐β1 production 67% based on prior literature. Both manipulations resulted in increased inflammation (elevated neutrophils, TNF, and tissue damage) and delayed healing (reduced TGF‐β1 and collagen). Our ABM reproduced the therapeutic effect of platelet‐derived growth factor/platelet releasate treatment as well as DFU debridement. We next simulated the expected effect of administering (1) a neutralizing anti‐TNF antibody, (2) an agent that would increase the activation of endogenous latent TGF‐β1, or (3) latent TGF‐β1 (which has a longer half‐life than active TGF‐β1), and found that these therapies would have similar effects regardless of the initial assumption of the derangement that underlies DFU (elevated TNF vs. reduced TGF‐β1). In silico methods may elucidate mechanisms of and suggest therapies for aberrant skin healing.


PLOS ONE | 2008

A Patient-Specific in silico Model of Inflammation and Healing Tested in Acute Vocal Fold Injury

Nicole Y. K. Li; Katherine Verdolini; Gilles Clermont; Qi Mi; Elaine N. Rubinstein; Patricia A. Hebda; Yoram Vodovotz

The development of personalized medicine is a primary objective of the medical community and increasingly also of funding and registration agencies. Modeling is generally perceived as a key enabling tool to target this goal. Agent-Based Models (ABMs) have previously been used to simulate inflammation at various scales up to the whole-organism level. We extended this approach to the case of a novel, patient-specific ABM that we generated for vocal fold inflammation, with the ultimate goal of identifying individually optimized treatments. ABM simulations reproduced trajectories of inflammatory mediators in laryngeal secretions of individuals subjected to experimental phonotrauma up to 4 hrs post-injury, and predicted the levels of inflammatory mediators 24 hrs post-injury. Subject-specific simulations also predicted different outcomes from behavioral treatment regimens to which subjects had not been exposed. We propose that this translational application of computational modeling could be used to design patient-specific therapies for the larynx, and will serve as a paradigm for future extension to other clinical domains.


Biophysical Journal | 2011

Continuum Model of Collective Cell Migration in Wound Healing and Colony Expansion

Julia Arciero; Qi Mi; Maria F. Branca; David J. Hackam; David Swigon

Collective cell migration plays an important role during wound healing and embryo development. Although the exact mechanisms that coordinate such migration are still unknown, experimental studies of moving cell layers have shown that the primary interactions governing the motion of the layer are the force of lamellipodia, the adhesion of cells to the substrate, and the adhesion of cells to each other. Here, we derive a two-dimensional continuum mechanical model of cell-layer migration that is based on a novel assumption of elastic deformation of the layer and incorporates basic mechanical interactions of cells as well as cell proliferation and apoptosis. The evolution equations are solved numerically using a level set method. The model successfully reproduces data from two types of experiments: 1), the contraction of an enterocyte cell layer during wound healing; and 2), the expansion of a radially symmetric colony of MDCK cells, both in the edge migration velocity and in cell-layer density. In accord with experimental observations, and in contrast to reaction-diffusion models, this model predicts a partial wound closure if lamellipod formation is inhibited at the wound edge and gives implications of the effect of spatially restricted proliferation.


Immunopharmacology and Immunotoxicology | 2010

Translational systems approaches to the biology of inflammation and healing.

Yoram Vodovotz; Gregory M. Constantine; James R. Faeder; Qi Mi; Jonathan E. Rubin; John Bartels; Joydeep Sarkar; Robert H. Squires; David O. Okonkwo; Jörg C. Gerlach; Ruben Zamora; Shirley Luckhart; Bard Ermentrout; Gary An

Inflammation is a complex, non-linear process central to many of the diseases that affect both developed and emerging nations. A systems-based understanding of inflammation, coupled to translational applications, is therefore necessary for efficient development of drugs and devices, for streamlining analyses at the level of populations, and for the implementation of personalized medicine. We have carried out an iterative and ongoing program of literature analysis, generation of prospective data, data analysis, and computational modeling in various experimental and clinical inflammatory disease settings. These simulations have been used to gain basic insights into the inflammatory response under baseline, gene-knockout, and drug-treated experimental animals for in silico studies associated with the clinical settings of sepsis, trauma, acute liver failure, and wound healing to create patient-specific simulations in polytrauma, traumatic brain injury, and vocal fold inflammation; and to gain insight into host-pathogen interactions in malaria, necrotizing enterocolitis, and sepsis. These simulations have converged with other systems biology approaches (e.g., functional genomics) to aid in the design of new drugs or devices geared towards modulating inflammation. Since they include both circulating and tissue-level inflammatory mediators, these simulations transcend typical cytokine networks by associating inflammatory processes with tissue/organ impacts via tissue damage/dysfunction. This framework has now allowed us to suggest how to modulate acute inflammation in a rational, individually optimized fashion. This plethora of computational and intertwined experimental/engineering approaches is the cornerstone of Translational Systems Biology approaches for inflammatory diseases.


Critical Care Medicine | 2012

A Two-Compartment Mathematical Model of Endotoxin-induced Inflammatory and Physiologic Alterations in Swine

Gary Nieman; David L. Brown; Joydeep Sarkar; Brian D. Kubiak; Cordelia Ziraldo; Joyeeta Dutta-Moscato; Christopher J. Vieau; Derek Barclay; Louis A. Gatto; Kristopher G. Maier; Gregory M. Constantine; Timothy R. Billiar; Ruben Zamora; Qi Mi; Steve Chang; Yoram Vodovotz

Objective:To gain insights into individual variations in acute inflammation and physiology. Design:Large-animal study combined with mathematical modeling. Setting:Academic large-animal and computational laboratories. Subjects:Outbred juvenile swine. Interventions:Four swine were instrumented and subjected to endotoxemia (100 µg/kg), followed by serial plasma sampling. Measurements and Main Results:Swine exhibited various degrees of inflammation and acute lung injury, including one death with severe acute lung injury (PaO2/FIO2 ratio &mgr;200 and static compliance &mgr;10 L/cm H2O). Plasma interleukin-1&bgr;, interleukin-4, interleukin-6, interleukin-8, interleukin-10, tumor necrosis factor-&agr;, high mobility group box-1, and NO2-/NO3- were significantly (p &mgr; .05) elevated over the course of the experiment. Principal component analysis was used to suggest principal drivers of inflammation. Based in part on principal component analysis, an ordinary differential equation model was constructed, consisting of the lung and the blood (as a surrogate for the rest of the body), in which endotoxin induces tumor necrosis factor-&agr; in monocytes in the blood, followed by the trafficking of these cells into the lung leading to the release of high mobility group box-1, which in turn stimulates the release of interleukin-1&bgr; from resident macrophages. The ordinary differential equation model also included blood pressure, PaO2, and FIO2, and a damage variable that summarizes the health of the animal. This ordinary differential equation model could be fit to both inflammatory and physiologic data in the individual swine. The predicted time course of damage could be matched to the oxygen index in three of the four swine. Conclusions:The approach described herein may aid in predicting inflammation and physiologic dysfunction in small cohorts of subjects with diverse phenotypes and outcomes.


PLOS ONE | 2011

A dynamic view of trauma/hemorrhage-induced inflammation in mice: principal drivers and networks.

Qi Mi; Gregory M. Constantine; Cordelia Ziraldo; Alexey Solovyev; Andres Torres; Rajaie Namas; Timothy Bentley; Timothy R. Billiar; Ruben Zamora; Juan Carlos Puyana; Yoram Vodovotz

Background Complex biological processes such as acute inflammation induced by trauma/hemorrhagic shock/ (T/HS) are dynamic and multi-dimensional. We utilized multiplexing cytokine analysis coupled with data-driven modeling to gain a systems perspective into T/HS. Methodology/Principal Findings Mice were subjected to surgical cannulation trauma (ST) ± hemorrhagic shock (HS; 25 mmHg), and followed for 1, 2, 3, or 4 h in each case. Serum was assayed for 20 cytokines and NO2 −/NO3 −. These data were analyzed using four data-driven methods (Hierarchical Clustering Analysis [HCA], multivariate analysis [MA], Principal Component Analysis [PCA], and Dynamic Network Analysis [DyNA]). Using HCA, animals subjected to ST vs. ST + HS could be partially segregated based on inflammatory mediator profiles, despite a large overlap. Based on MA, interleukin [IL]-12p40/p70 (IL-12.total), monokine induced by interferon-γ (CXCL-9) [MIG], and IP-10 were the best discriminators between ST and ST/HS. PCA suggested that the inflammatory mediators found in the three main principal components in animals subjected to ST were IL-6, IL-10, and IL-13, while the three principal components in ST + HS included a large number of cytokines including IL-6, IL-10, keratinocyte-derived cytokine (CXCL-1) [KC], and tumor necrosis factor-α [TNF-α]. DyNA suggested that the circulating mediators produced in response to ST were characterized by a high degree of interconnection/complexity at all time points; the response to ST + HS consisted of different central nodes, and exhibited zero network density over the first 2 h with lesser connectivity vs. ST at all time points. DyNA also helped link the conclusions from MA and PCA, in that central nodes consisting of IP-10 and IL-12 were seen in ST, while MIG and IL-6 were central nodes in ST + HS. Conclusions/Significance These studies help elucidate the dynamics of T/HS-induced inflammation, complementing other forms of dynamic mechanistic modeling. These methods should be applicable to the analysis of other complex biological processes.


Annals of Surgery | 2016

Temporal Patterns of Circulating Inflammation Biomarker Networks Differentiate Susceptibility to Nosocomial Infection Following Blunt Trauma in Humans.

Rami A. Namas; Yoram Vodovotz; Khalid Almahmoud; Othman Abdul-Malak; Akram Zaaqoq; Rajaie Namas; Qi Mi; Derek Barclay; Brian S. Zuckerbraun; Andrew B. Peitzman; Jason L. Sperry; Timothy R. Billiar

BACKGROUND Severe traumatic injury can lead to immune dysfunction that renders trauma patients susceptible to nosocomial infections (NI) and prolonged intensive care unit (ICU) stays. We hypothesized that early circulating biomarker patterns following trauma would correlate with sustained immune dysregulation associated with NI and remote organ failure. METHODS In a cohort of 472 blunt trauma survivors studied over an 8-year period, 127 patients (27%) were diagnosed with NI versus 345 trauma patients without NI. To perform a pairwise, case-control study with 1:1 matching, 44 of the NI patients were compared with 44 no-NI trauma patients selected by matching patient demographics and injury characteristics. Plasma obtained upon admission and over time were assayed for 26 inflammatory mediators and analyzed for the presence of dynamic networks. RESULTS Significant differences in ICU length of stay (LOS), hospital LOS, and days on mechanical ventilation were observed in the NI patients versus no-NI patients. Although NI was not detected until day 7, multiple mediators were significantly elevated within the first 24 hours in patients who developed NI. Circulating inflammation biomarkers exhibited 4 distinct dynamic patterns, of which 2 clearly distinguish patients destined to develop NI from those who did not. Mediator network connectivity analysis revealed a higher, coordinated degree of activation of both innate and lymphoid pathways in the NI patients over the initial 24 hours. CONCLUSIONS These studies implicate unique dynamic immune responses, reflected in circulating biomarkers that differentiate patients prone to persistent critical illness and infections following injury, independent of mechanism of injury, injury severity, age, or sex.


PLOS ONE | 2013

Central Role for MCP-1/CCL2 in Injury-Induced Inflammation Revealed by In Vitro , In Silico , and Clinical Studies

Cordelia Ziraldo; Yoram Vodovotz; Rami A. Namas; Khalid Almahmoud; Victor Tapias; Qi Mi; Derek Barclay; Bahiyyah S. Jefferson; Guoqiang Chen; Timothy R. Billiar; Ruben Zamora

The translation of in vitro findings to clinical outcomes is often elusive. Trauma/hemorrhagic shock (T/HS) results in hepatic hypoxia that drives inflammation. We hypothesize that in silico methods would help bridge in vitro hepatocyte data and clinical T/HS, in which the liver is a primary site of inflammation. Primary mouse hepatocytes were cultured under hypoxia (1% O2) or normoxia (21% O2) for 1–72 h, and both the cell supernatants and protein lysates were assayed for 18 inflammatory mediators by Luminex™ technology. Statistical analysis and data-driven modeling were employed to characterize the main components of the cellular response. Statistical analyses, hierarchical and k-means clustering, Principal Component Analysis, and Dynamic Network Analysis suggested MCP-1/CCL2 and IL-1α as central coordinators of hepatocyte-mediated inflammation in C57BL/6 mouse hepatocytes. Hepatocytes from MCP-1-null mice had altered dynamic inflammatory networks. Circulating MCP-1 levels segregated human T/HS survivors from non-survivors. Furthermore, T/HS survivors with elevated early levels of plasma MCP-1 post-injury had longer total lengths of stay, longer intensive care unit lengths of stay, and prolonged requirement for mechanical ventilation vs. those with low plasma MCP-1. This study identifies MCP-1 as a main driver of the response of hepatocytes in vitro and as a biomarker for clinical outcomes in T/HS, and suggests an experimental and computational framework for discovery of novel clinical biomarkers in inflammatory diseases.


Molecular Medicine | 2012

Hemoadsorption reprograms inflammation in experimental gram-negative septic peritonitis: insights from in vivo and in silico studies.

Rami A. Namas; Rajaie Namas; Claudio Lagoa; Derek Barclay; Qi Mi; Ruben Zamora; Zhi-Yong Peng; Morgan V. Fedorchak; Isabella E. Valenti; William J. Federspiel; John A. Kellum; Yoram Vodovotz

Improper compartmentalization of the inflammatory response leads to systemic inflammation in sepsis. Hemoadsorption (HA) is an emerging approach to modulate sepsis-induced inflammation. We sought to define the effects of HA on inflammatory compartmentalization in Escherichia coli-induced fibrin peritonitis in rats. Hypothesis: HA both reprograms and recompartmentalizes inflammation in sepsis. Sprague Dawley male rats were subjected to E. coli peritonitis and, after 24 h, were randomized to HA or sham treatment (sepsis alone). Venous blood samples collected at 0, 1, 3 and 6 h (that is, 24–30 h of total experimental sepsis), and peritoneal samples collected at 0 and 6 h, were assayed for 14 cytokines along with NO2−/NO3−. Bacterial counts were assessed in the peritoneal fluid at 0 and 6 h. Plasma tumor necrosis factor (TNF)-α, interleukin (IL)-6, CXCL-1, and CCL2 were significantly reduced in HA versus sham. Principal component analysis (PCA) suggested that inflammation in sham was driven by IL-6 and TNF-α, whereas HA-associated inflammation was driven primarily by TNF-α, CXCL-1, IL-10 and CCL2. Whereas peritoneal bacterial counts, plasma aspartate transaminase levels and peritoneal IL-5, IL-6, IL-18, interferon (IFN)-γ and NO2−/NO3− were significantly lower, both CXCL-1 and CCL2 as well as the peritoneal-to-plasma ratios of TNF-α, CXCL-1 and CCL2 were significantly higher in HA versus sham, suggesting that HA-induced inflammatory recompartmentalization leads to the different inflammatory drivers discerned in part by PCA. In conclusion, this study demonstrates the utility of combined in vivo/in silico methods and suggests that HA exerts differential effects on mediator gradients between local and systemic compartments that ultimately benefit the host.

Collaboration


Dive into the Qi Mi's collaboration.

Top Co-Authors

Avatar

Yoram Vodovotz

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Ruben Zamora

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Derek Barclay

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rajaie Namas

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Rami A. Namas

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge