Skip to main content

Chen Laboratory

The Chen lab develops novel imaging methods to understand mechanisms of cardiovascular disease, to image disease as it progresses and to improve cardiovascular outcomes by targeting or monitoring therapies using molecular imaging. He is applying these novel technologies to heart failure and cardio-oncology.

Current projects include:

Molecular imaging of cardiac autophagy in vivo

Molecular imaging of cell death, namely autophagy, necrosis and apoptosis, relies on intravenously injectable contrast agents to target specific biological states and provide sensitive readout. The ability to image cell death processes noninvasively in vivo allows the kinetics to be studied during disease progression and regression. Autophagy is a conserved catabolic process where cells recycle part of the cytoplasm during stress. Interestingly, autophagy can exert either detrimental or protective pathophysiological effects on the heart during disease. We demonstrated for the first time probe-based imaging of autophagy in a beating mouse heart. In mouse hearts with ischemia-reperfusion injury (IR), autophagy induction by Rapamycin (RAP) was visualized and quantified (Figure 1).

 

MCRI Chen
Figure 1. Visualizing in vivo cardiac autophagy. (A-D) 3D tomographic fluorescence imaging with an autophagy-sensitive probe in Rapamycin (RAP) treated, ischemia-reperfusion (IR) injured mouse heart. Probe activation is localized to the anterior lateral wall of the heart, consistent with the site of ischemic injury. The fluorescence signal was fused with Computed Tomography (CT) for anatomical coregistration and accurate volumetric quantification (E). n=7 in each cohort, Student’s t-test.

 

The Autophagy imaging probe mechanism is further elucidated in cell culture (Figure 2). Probe activation following autophagy induction is three-fold: i) the rapid translocation of the autophagy probe into the cytoplasm, ii) the high degree of co-trafficking between the autophagy probe and early autophagosomes and iii) the activatable nature of the autophagy probe upon entering autophagolysosomes. We hypothesize that bioconjugation chemistry strategies can be applied to engineer second-generation autophagy sensing probes with enhanced sensitivity and specificity to monitor autophagy levels non-invasively in vivo.

Chen Lab Figure
 Figure 2. In H9C2 cardiomyocytes starved for 24 hours to induce autophagy, Autophagy Detecting Nanoparticle (ADN) visualized by DX-1 antibody staining of dextran is seen diffusely throughout the cytoplasm within 15 minutes (A). By 60 minutes, ADN is seen colocalizing with early autophagosomes labeled by LC3-GFP (B). After 4 hours of probe incubation, ADN fluorescence is seen localized with autophagolysosomes marked by LC3-RFP (C). (D). ADN accumulation visualized by electron microscopy shows double-membraned autophagosomes loaded with electron-dense puncta, which is not seen in controls without ADN added. The findings are seen consistently in 3-7 biological and technical replicates. (A-C): Scale bar=20μm.
Theranostic approaches with rationally designed nanoprobes

Theranostics are imaging agents with dual therapeutic and diagnostic capabilities. Developing theranostics would allow us to see disease processes non-invasively in the body, simultaneously applying therapeutics at the site of injury, and yield real-time readout of response to treatment. An example of a theranostic nanoprobe is a biocompatible polymer construct engineered with nucleic acid chelating fluorochromes. The probe targets nucleic acids released during cell rupture to facilitate imaging of cardiomyocyte necrosis (Figure 3).

MCRI Chen 3
Figure 3. Necrosis-sensing optical nanoprobe. (A) Synthesis of the active (Dex-TO) and control (Dex-Cy5.5) probes. (B) Multivalent Dex-TO fluorescence increases robustly with DNA. (C) Dex-TO is excluded from healthy cells, and specifically taken up by necrotic cells. (D) In infarcted mouse heart slices, Dex-TO fluorescence correlates with TTC staining of tissue necrosis. No correlation is seen with control Dex-Cy5.5. (E) Receiver Operating Characteristics of Dex-TO (Area Under the Curve=0.88, n=7 mice).

 

Extracellular nucleic acids are robust proinflammatory molecules that elicit immune cell infiltration. Theranostic nucleic acid binding nanoprobe developed has nanomolar affinity for mammalian and bacterial nucleic acids and attenuates the production of inflammatory cytokines from activated macrophages exposed to DNA or RNA (Figure 4). In mice treated with the nanoprobe, inflammation was reduced at 24 hours after myocardial ischemia-reperfusion injury, leading to improved cardiac healing by day 7. Food and Drug Administration (FDA)-approved biocompatible polymer platform further allows pharmacokinetics to be optimized to enhance diagnostic and therapeutic efficacies and can be translatable from molecular targeting to improve treatment outcomes.

MCRI Chen 4
Figure 4. Schematics of extracellular nucleic acid chelating nanoprobe to exert anti-inflammatory and cardioprotective effects.
people

Lab members

  • Asma Boukhalfa, Postdoctoral Fellow

 

Previous members

  • Andrew Kung, Undergraduate Researcher
  • Ada Yu, Undergraduate Researcher
  • Fernanda Garcia, Undergraduate Researcher
Contact info
user
Howard Chen, PhD
Jump back to top