Network


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

Hotspot


Dive into the research topics where Chad M. Trent is active.

Publication


Featured researches published by Chad M. Trent.


Cell Metabolism | 2012

Lipid Metabolism and Toxicity in the Heart

Ira J. Goldberg; Chad M. Trent; P. Christian Schulze

The heart has both the greatest caloric needs and the most robust oxidation of fatty acids (FAs). Under pathological conditions such as obesity and type 2 diabetes, cardiac uptake and oxidation are not balanced and hearts accumulate lipid potentially leading to cardiac lipotoxicity. We will first review the pathways utilized by the heart to acquire FAs from the circulation and to store triglyceride intracellularly. Then we will describe mouse models in which excess lipid accumulation causes heart dysfunction and experiments performed to alleviate this toxicity. Finally, the known relationships between heart lipid metabolism and dysfunction in humans will be summarized.


Cell Metabolism | 2014

High Mobility Group Box 1 is Dispensable for Autophagy, Mitochondrial Quality Control and Organ Function in Vivo

Peter Huebener; Geum-Youn Gwak; Jean Philippe Pradere; Catarina M. Quinzii; Richard A. Friedman; Chyuan Sheng Lin; Chad M. Trent; Ingmar Mederacke; Enpeng Zhao; Yuxi Lin; Ira J. Goldberg; Mark J. Czaja; Robert F. Schwabe

In vitro studies have demonstrated a critical role for high-mobility group box 1 (HMGB1) in autophagy and the autophagic clearance of dysfunctional mitochondria, resulting in severe mitochondrial fragmentation and profound disturbances of mitochondrial respiration in HMGB1-deficient cells. Here, we investigated the effects of HMGB1 deficiency on autophagy and mitochondrial function in vivo, using conditional Hmgb1 ablation in the liver and heart. Unexpectedly, deletion of Hmgb1 in hepatocytes or cardiomyocytes, two cell types with abundant mitochondria, did not alter mitochondrial structure or function, organ function, or long-term survival. Moreover, hepatic autophagy and mitophagy occurred normally in the absence of Hmgb1, and absence of Hmgb1 did not significantly affect baseline and glucocorticoid-induced hepatic gene expression. Collectively, our findings suggest that HMGB1 is dispensable for autophagy, mitochondrial quality control, the regulation of gene expression, and organ function in the adult organism.


Circulation-heart Failure | 2013

Peroxisome Proliferator–Activated Receptor-γ Activation Prevents Sepsis-Related Cardiac Dysfunction and Mortality In Mice

Konstantinos Drosatos; Raffay Khan; Chad M. Trent; Hongfeng Jiang; Ni-Huiping Son; William S. Blaner; Shunichi Homma; P. Christian Schulze; Ira J. Goldberg

Impaired cardiac contractility contributes to the hypotension and increased mortality that occur with sepsis1. A possible cause of sepsis-mediated cardiac dysfunction is reduced energy production due in part to compromised fatty acid oxidation (FAO)2-5 and glucose catabolism3, 6. Thus, it is likely that sepsis compromises cardiac energy production, which might be the major cause of cardiac dysfunction. Alternatively, sepsis induces the production of inflammatory cytokines, such as tumor necrosis factor (TNF) α, interleukin (IL)-1 and IL-6, and these might directly alter heart function7-9. Intraperitoneal (i.p.) injection of lipopolysaccharide (LPS) has been extensively used to model many of the clinical features of sepsis, including elevated inflammation and cardiac dysfunction10. LPS leads to production of inflammatory cytokines7-9, 11 and also reduces cardiac energy utilization2, 3, 12. Nuclear receptors, particularly peroxisomal proliferator-activated receptors (PPARs), regulate cardiac FAO. The PPAR family consists of three members, PPARα, PPARδ and PPARγ. PPARα increases FA storage in triglycerides13 and FAO in heart14 and induces expression of peroxisomal and mitochondrial enzymes. Besides PPARα, cardiac FAO can be increased by activation of PPARγ15 or PPARδ16. Cardiomyocyte-specific overexpression of PPARα14 or PPARγ17 leads to cardiac lipid accumulation, an indication that lipid uptake exceeds FAO. PPARγ-coactivator-1 (PGC-1) α and β18 enhance FAO and mitochondrial biogenesis19. Both PPARα and PGC-1 mRNA levels are markedly reduced in the heart by LPS administration2, 3, 12, 20, while PPARγ is not affected2. Our group showed that maintenance of normal cardiac FAO via c-Jun-N-terminal kinase (JNK) inhibitor-mediated prevention of PPARα downregulation rescued cardiac function in septic mice despite elevated expression of cardiac inflammatory markers. In a similar context constitutive cardiac expression of PGC-1β prevented cardiac dysfunction that was caused by LPS-mediated sepsis3, an observation that was proposed to be due to improvement in cardiac FAO and attenuation of reactive oxygen species production. In the current study we show that constitutive cardiomyocyte-specific expression of PPARγ or systemic administration of the PPARγ agonist, rosiglitazone, increased cardiac FAO and prevented cardiac dysfunction in mice with LPS-induced sepsis, despite increased expression of cardiac inflammatory markers. In addition, we show that rosiglitazone-mediated activation of PPARγ prevents the loss of cardiac mitochondria that occurs in sepsis. Moreover, we show that restoration of cardiac FAO by rosiglitazone not only prevents but also treats LPS-induced heart dysfunction and improves survival. Thus the use of rosiglitazone is proposed as a potential treatment for septic cardiac dysfunction.Background—Cardiac dysfunction with sepsis is associated with both inflammation and reduced fatty acid oxidation. We hypothesized that energy deprivation accounts for sepsis-related cardiac dysfunction. Methods and Results—Escherichia coli lipopolysaccharide (LPS) administered to C57BL/6 mice (wild type) induced cardiac dysfunction and reduced fatty acid oxidation and mRNA levels of peroxisome proliferator–activated receptor (PPAR)-&agr; and its downstream targets within 6–8 hours. Transgenic mice in which cardiomyocyte-specific expression of PPAR&ggr; is driven by the &agr;-myosin heavy chain promoter (&agr;MHC-PPAR&ggr;) were protected from LPS-induced cardiac dysfunction. Despite a reduction in PPAR&agr;, fatty acid oxidation and associated genes were not decreased in hearts of LPS-treated &agr;MHC-PPAR&ggr; mice. LPS treatment, however, continued to induce inflammation-related genes, such as interleukin-1&agr;, interleukin-1&bgr;, interleukin-6, and tumor necrosis factor-&agr; in hearts of &agr;MHC-PPAR&ggr; mice. Treatment of wild-type mice with LPS and the PPAR&ggr; agonist, rosiglitazone, but not the PPAR&agr; agonist (WY-14643), increased fatty acid oxidation, prevented LPS-mediated reduction of mitochondria, and treated cardiac dysfunction, as well as it improved survival, despite continued increases in the expression of cardiac inflammatory markers. Conclusions—Activation of PPAR&ggr; in LPS-treated mice prevented cardiac dysfunction and mortality, despite development of cardiac inflammation and PPAR&agr; downregulation.


Journal of Lipid Research | 2014

Lipoprotein lipase activity is required for cardiac lipid droplet production

Chad M. Trent; Shuiqing Yu; Yunying Hu; Nathan Skoller; Lesley Ann Huggins; Shunichi Homma; Ira J. Goldberg

The rodent heart accumulates TGs and lipid droplets during fasting. The sources of heart lipids could be either FFAs liberated from adipose tissue or FAs from lipoprotein-associated TGs via the action of lipoprotein lipase (LpL). Because circulating levels of FFAs increase during fasting, it has been assumed that albumin transported FFAs are the source of lipids within heart lipid droplets. We studied mice with three genetic mutations: peroxisomal proliferator-activated receptor α deficiency, cluster of differentiation 36 (CD36) deficiency, and heart-specific LpL deletion. All three genetically altered groups of mice had defective accumulation of lipid droplet TGs. Moreover, hearts from mice treated with poloxamer 407, an inhibitor of lipoprotein TG lipolysis, also failed to accumulate TGs, despite increased uptake of FFAs. TG storage did not impair maximal cardiac function as measured by stress echocardiography. Thus, LpL hydrolysis of circulating lipoproteins is required for the accumulation of lipids in the heart of fasting mice.


Circulation-heart Failure | 2013

Peroxisome Proliferator–Activated Receptor-γ Activation Prevents Sepsis-Related Cardiac Dysfunction and Mortality In MiceClinical Perspective

Konstantinos Drosatos; Raffay Khan; Chad M. Trent; Hongfeng Jiang; Ni-Huiping Son; William S. Blaner; Shunichi Homma; P. Christian Schulze; Ira J. Goldberg

Impaired cardiac contractility contributes to the hypotension and increased mortality that occur with sepsis1. A possible cause of sepsis-mediated cardiac dysfunction is reduced energy production due in part to compromised fatty acid oxidation (FAO)2-5 and glucose catabolism3, 6. Thus, it is likely that sepsis compromises cardiac energy production, which might be the major cause of cardiac dysfunction. Alternatively, sepsis induces the production of inflammatory cytokines, such as tumor necrosis factor (TNF) α, interleukin (IL)-1 and IL-6, and these might directly alter heart function7-9. Intraperitoneal (i.p.) injection of lipopolysaccharide (LPS) has been extensively used to model many of the clinical features of sepsis, including elevated inflammation and cardiac dysfunction10. LPS leads to production of inflammatory cytokines7-9, 11 and also reduces cardiac energy utilization2, 3, 12. Nuclear receptors, particularly peroxisomal proliferator-activated receptors (PPARs), regulate cardiac FAO. The PPAR family consists of three members, PPARα, PPARδ and PPARγ. PPARα increases FA storage in triglycerides13 and FAO in heart14 and induces expression of peroxisomal and mitochondrial enzymes. Besides PPARα, cardiac FAO can be increased by activation of PPARγ15 or PPARδ16. Cardiomyocyte-specific overexpression of PPARα14 or PPARγ17 leads to cardiac lipid accumulation, an indication that lipid uptake exceeds FAO. PPARγ-coactivator-1 (PGC-1) α and β18 enhance FAO and mitochondrial biogenesis19. Both PPARα and PGC-1 mRNA levels are markedly reduced in the heart by LPS administration2, 3, 12, 20, while PPARγ is not affected2. Our group showed that maintenance of normal cardiac FAO via c-Jun-N-terminal kinase (JNK) inhibitor-mediated prevention of PPARα downregulation rescued cardiac function in septic mice despite elevated expression of cardiac inflammatory markers. In a similar context constitutive cardiac expression of PGC-1β prevented cardiac dysfunction that was caused by LPS-mediated sepsis3, an observation that was proposed to be due to improvement in cardiac FAO and attenuation of reactive oxygen species production. In the current study we show that constitutive cardiomyocyte-specific expression of PPARγ or systemic administration of the PPARγ agonist, rosiglitazone, increased cardiac FAO and prevented cardiac dysfunction in mice with LPS-induced sepsis, despite increased expression of cardiac inflammatory markers. In addition, we show that rosiglitazone-mediated activation of PPARγ prevents the loss of cardiac mitochondria that occurs in sepsis. Moreover, we show that restoration of cardiac FAO by rosiglitazone not only prevents but also treats LPS-induced heart dysfunction and improves survival. Thus the use of rosiglitazone is proposed as a potential treatment for septic cardiac dysfunction.Background—Cardiac dysfunction with sepsis is associated with both inflammation and reduced fatty acid oxidation. We hypothesized that energy deprivation accounts for sepsis-related cardiac dysfunction. Methods and Results—Escherichia coli lipopolysaccharide (LPS) administered to C57BL/6 mice (wild type) induced cardiac dysfunction and reduced fatty acid oxidation and mRNA levels of peroxisome proliferator–activated receptor (PPAR)-&agr; and its downstream targets within 6–8 hours. Transgenic mice in which cardiomyocyte-specific expression of PPAR&ggr; is driven by the &agr;-myosin heavy chain promoter (&agr;MHC-PPAR&ggr;) were protected from LPS-induced cardiac dysfunction. Despite a reduction in PPAR&agr;, fatty acid oxidation and associated genes were not decreased in hearts of LPS-treated &agr;MHC-PPAR&ggr; mice. LPS treatment, however, continued to induce inflammation-related genes, such as interleukin-1&agr;, interleukin-1&bgr;, interleukin-6, and tumor necrosis factor-&agr; in hearts of &agr;MHC-PPAR&ggr; mice. Treatment of wild-type mice with LPS and the PPAR&ggr; agonist, rosiglitazone, but not the PPAR&agr; agonist (WY-14643), increased fatty acid oxidation, prevented LPS-mediated reduction of mitochondria, and treated cardiac dysfunction, as well as it improved survival, despite continued increases in the expression of cardiac inflammatory markers. Conclusions—Activation of PPAR&ggr; in LPS-treated mice prevented cardiac dysfunction and mortality, despite development of cardiac inflammation and PPAR&agr; downregulation.


Heart Rhythm | 2016

Mitochondrial oxidative stress during cardiac lipid overload causes intracellular calcium leak and arrhythmia

Leroy C. Joseph; Prakash Subramanyam; Christopher Radlicz; Chad M. Trent; Vivek Iyer; Henry M. Colecraft; John P. Morrow

BACKGROUND Diabetes and obesity are associated with an increased risk of arrhythmia and sudden cardiac death. Abnormal lipid accumulation is observed in cardiomyocytes of obese and diabetic patients, which may contribute to arrhythmia, but the mechanisms are poorly understood. A transgenic mouse model of cardiac lipid overload, the peroxisome proliferator-activated receptor-γ (PPARg) cardiac overexpression mouse, has long QT and increased ventricular ectopy. OBJECTIVE The purpose of this study was to evaluate the hypothesis that the increase in ventricular ectopy during cardiac lipid overload is caused by abnormalities in calcium handling due to increased mitochondrial oxidative stress. METHODS Ventricular myocytes were isolated from adult mouse hearts to record sparks and calcium transients. Mice were implanted with heart rhythm monitors for in vivo recordings. RESULTS PPARg cardiomyocytes have more frequent triggered activity and increased sparks compared to control. Sparks and triggered activity are reduced by mitotempo, a mitochondrial-targeted antioxidant. This is explained by a significant increase in oxidation of RyR2. Calcium transients are increased in amplitude, and sarcoplasmic reticulum (SR) calcium stores are increased in PPARg cardiomyocytes. Computer modeling of the cardiac action potential demonstrates that long QT contributes to increased SR calcium. Mitotempo decreased ventricular ectopy in vivo. CONCLUSION During cardiac lipid overload, mitochondrial oxidative stress causes increased SR calcium leak by oxidizing RyR2 channels. This promotes ventricular ectopy, which is significantly reduced in vivo by a mitochondrial-targeted antioxidant. These results suggest a potential role for mitochondrial-targeted antioxidants in preventing arrhythmia and sudden cardiac death in obese and diabetic patients.


Circulation-heart Failure | 2013

Peroxisome Proliferator-Activated Receptor- Activation Prevents Sepsis-Related Cardiac Dysfunction and Mortality In Mice

Konstantinos Drosatos; Raffay Khan; Chad M. Trent; Hongfeng Jiang; Ni-Huiping Son; William S. Blaner; Shunichi Homma; P. C. Schulze; Ira J. Goldberg

Impaired cardiac contractility contributes to the hypotension and increased mortality that occur with sepsis1. A possible cause of sepsis-mediated cardiac dysfunction is reduced energy production due in part to compromised fatty acid oxidation (FAO)2-5 and glucose catabolism3, 6. Thus, it is likely that sepsis compromises cardiac energy production, which might be the major cause of cardiac dysfunction. Alternatively, sepsis induces the production of inflammatory cytokines, such as tumor necrosis factor (TNF) α, interleukin (IL)-1 and IL-6, and these might directly alter heart function7-9. Intraperitoneal (i.p.) injection of lipopolysaccharide (LPS) has been extensively used to model many of the clinical features of sepsis, including elevated inflammation and cardiac dysfunction10. LPS leads to production of inflammatory cytokines7-9, 11 and also reduces cardiac energy utilization2, 3, 12. Nuclear receptors, particularly peroxisomal proliferator-activated receptors (PPARs), regulate cardiac FAO. The PPAR family consists of three members, PPARα, PPARδ and PPARγ. PPARα increases FA storage in triglycerides13 and FAO in heart14 and induces expression of peroxisomal and mitochondrial enzymes. Besides PPARα, cardiac FAO can be increased by activation of PPARγ15 or PPARδ16. Cardiomyocyte-specific overexpression of PPARα14 or PPARγ17 leads to cardiac lipid accumulation, an indication that lipid uptake exceeds FAO. PPARγ-coactivator-1 (PGC-1) α and β18 enhance FAO and mitochondrial biogenesis19. Both PPARα and PGC-1 mRNA levels are markedly reduced in the heart by LPS administration2, 3, 12, 20, while PPARγ is not affected2. Our group showed that maintenance of normal cardiac FAO via c-Jun-N-terminal kinase (JNK) inhibitor-mediated prevention of PPARα downregulation rescued cardiac function in septic mice despite elevated expression of cardiac inflammatory markers. In a similar context constitutive cardiac expression of PGC-1β prevented cardiac dysfunction that was caused by LPS-mediated sepsis3, an observation that was proposed to be due to improvement in cardiac FAO and attenuation of reactive oxygen species production. In the current study we show that constitutive cardiomyocyte-specific expression of PPARγ or systemic administration of the PPARγ agonist, rosiglitazone, increased cardiac FAO and prevented cardiac dysfunction in mice with LPS-induced sepsis, despite increased expression of cardiac inflammatory markers. In addition, we show that rosiglitazone-mediated activation of PPARγ prevents the loss of cardiac mitochondria that occurs in sepsis. Moreover, we show that restoration of cardiac FAO by rosiglitazone not only prevents but also treats LPS-induced heart dysfunction and improves survival. Thus the use of rosiglitazone is proposed as a potential treatment for septic cardiac dysfunction.Background—Cardiac dysfunction with sepsis is associated with both inflammation and reduced fatty acid oxidation. We hypothesized that energy deprivation accounts for sepsis-related cardiac dysfunction. Methods and Results—Escherichia coli lipopolysaccharide (LPS) administered to C57BL/6 mice (wild type) induced cardiac dysfunction and reduced fatty acid oxidation and mRNA levels of peroxisome proliferator–activated receptor (PPAR)-&agr; and its downstream targets within 6–8 hours. Transgenic mice in which cardiomyocyte-specific expression of PPAR&ggr; is driven by the &agr;-myosin heavy chain promoter (&agr;MHC-PPAR&ggr;) were protected from LPS-induced cardiac dysfunction. Despite a reduction in PPAR&agr;, fatty acid oxidation and associated genes were not decreased in hearts of LPS-treated &agr;MHC-PPAR&ggr; mice. LPS treatment, however, continued to induce inflammation-related genes, such as interleukin-1&agr;, interleukin-1&bgr;, interleukin-6, and tumor necrosis factor-&agr; in hearts of &agr;MHC-PPAR&ggr; mice. Treatment of wild-type mice with LPS and the PPAR&ggr; agonist, rosiglitazone, but not the PPAR&agr; agonist (WY-14643), increased fatty acid oxidation, prevented LPS-mediated reduction of mitochondria, and treated cardiac dysfunction, as well as it improved survival, despite continued increases in the expression of cardiac inflammatory markers. Conclusions—Activation of PPAR&ggr; in LPS-treated mice prevented cardiac dysfunction and mortality, despite development of cardiac inflammation and PPAR&agr; downregulation.


Circulation-heart Failure | 2013

PPARγ Activation Prevents Sepsis-Related Cardiac Dysfunction and Mortality in Mice

Konstantinos Drosatos; Raffay Khan; Chad M. Trent; Hongfeng Jiang; Ni-Huiping Son; William S. Blaner; Shunichi Homma; P. Christian Schulze; Ira J. Goldberg

Impaired cardiac contractility contributes to the hypotension and increased mortality that occur with sepsis1. A possible cause of sepsis-mediated cardiac dysfunction is reduced energy production due in part to compromised fatty acid oxidation (FAO)2-5 and glucose catabolism3, 6. Thus, it is likely that sepsis compromises cardiac energy production, which might be the major cause of cardiac dysfunction. Alternatively, sepsis induces the production of inflammatory cytokines, such as tumor necrosis factor (TNF) α, interleukin (IL)-1 and IL-6, and these might directly alter heart function7-9. Intraperitoneal (i.p.) injection of lipopolysaccharide (LPS) has been extensively used to model many of the clinical features of sepsis, including elevated inflammation and cardiac dysfunction10. LPS leads to production of inflammatory cytokines7-9, 11 and also reduces cardiac energy utilization2, 3, 12. Nuclear receptors, particularly peroxisomal proliferator-activated receptors (PPARs), regulate cardiac FAO. The PPAR family consists of three members, PPARα, PPARδ and PPARγ. PPARα increases FA storage in triglycerides13 and FAO in heart14 and induces expression of peroxisomal and mitochondrial enzymes. Besides PPARα, cardiac FAO can be increased by activation of PPARγ15 or PPARδ16. Cardiomyocyte-specific overexpression of PPARα14 or PPARγ17 leads to cardiac lipid accumulation, an indication that lipid uptake exceeds FAO. PPARγ-coactivator-1 (PGC-1) α and β18 enhance FAO and mitochondrial biogenesis19. Both PPARα and PGC-1 mRNA levels are markedly reduced in the heart by LPS administration2, 3, 12, 20, while PPARγ is not affected2. Our group showed that maintenance of normal cardiac FAO via c-Jun-N-terminal kinase (JNK) inhibitor-mediated prevention of PPARα downregulation rescued cardiac function in septic mice despite elevated expression of cardiac inflammatory markers. In a similar context constitutive cardiac expression of PGC-1β prevented cardiac dysfunction that was caused by LPS-mediated sepsis3, an observation that was proposed to be due to improvement in cardiac FAO and attenuation of reactive oxygen species production. In the current study we show that constitutive cardiomyocyte-specific expression of PPARγ or systemic administration of the PPARγ agonist, rosiglitazone, increased cardiac FAO and prevented cardiac dysfunction in mice with LPS-induced sepsis, despite increased expression of cardiac inflammatory markers. In addition, we show that rosiglitazone-mediated activation of PPARγ prevents the loss of cardiac mitochondria that occurs in sepsis. Moreover, we show that restoration of cardiac FAO by rosiglitazone not only prevents but also treats LPS-induced heart dysfunction and improves survival. Thus the use of rosiglitazone is proposed as a potential treatment for septic cardiac dysfunction.Background—Cardiac dysfunction with sepsis is associated with both inflammation and reduced fatty acid oxidation. We hypothesized that energy deprivation accounts for sepsis-related cardiac dysfunction. Methods and Results—Escherichia coli lipopolysaccharide (LPS) administered to C57BL/6 mice (wild type) induced cardiac dysfunction and reduced fatty acid oxidation and mRNA levels of peroxisome proliferator–activated receptor (PPAR)-&agr; and its downstream targets within 6–8 hours. Transgenic mice in which cardiomyocyte-specific expression of PPAR&ggr; is driven by the &agr;-myosin heavy chain promoter (&agr;MHC-PPAR&ggr;) were protected from LPS-induced cardiac dysfunction. Despite a reduction in PPAR&agr;, fatty acid oxidation and associated genes were not decreased in hearts of LPS-treated &agr;MHC-PPAR&ggr; mice. LPS treatment, however, continued to induce inflammation-related genes, such as interleukin-1&agr;, interleukin-1&bgr;, interleukin-6, and tumor necrosis factor-&agr; in hearts of &agr;MHC-PPAR&ggr; mice. Treatment of wild-type mice with LPS and the PPAR&ggr; agonist, rosiglitazone, but not the PPAR&agr; agonist (WY-14643), increased fatty acid oxidation, prevented LPS-mediated reduction of mitochondria, and treated cardiac dysfunction, as well as it improved survival, despite continued increases in the expression of cardiac inflammatory markers. Conclusions—Activation of PPAR&ggr; in LPS-treated mice prevented cardiac dysfunction and mortality, despite development of cardiac inflammation and PPAR&agr; downregulation.


American Journal of Physiology-heart and Circulatory Physiology | 2014

Cardiac dysfunction in β-carotene-15,15′-dioxygenase-deficient mice is associated with altered retinoid and lipid metabolism

Seung-Ah Lee; Hongfeng Jiang; Chad M. Trent; Jason J. Yuen; Sureshbabu Narayanasamy; Robert W. Curley; Earl H. Harrison; Ira J. Goldberg; Mathew S. Maurer; William S. Blaner


Circulation Research | 2014

Abstract O-1: Klf5 Regulates Cardiac Pparα and Med13 and affects Cardiac Fatty Acid Metabolism And Obesity

Konstantinos Drosatos; Nina M. Pollak; Panagiotis Ntziachristos; Chad M. Trent; Yunying Hu; Shunichi Homma; Iannis Aifantis; Ira J. Goldberg

Collaboration


Dive into the Chad M. Trent's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge