Network


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

Hotspot


Dive into the research topics where Jon Sin is active.

Publication


Featured researches published by Jon Sin.


PLOS Pathogens | 2014

Coxsackievirus B exits the host cell in shed microvesicles displaying autophagosomal markers.

Scott M. Robinson; Ginger Tsueng; Jon Sin; Vrushali Mangale; Shahad Rahawi; Laura L. McIntyre; Wesley Williams; Nelson Kha; Casey Cruz; Bryan M. Hancock; David P. Nguyen; M. Richard Sayen; Brett J. Hilton; Kelly S. Doran; Anca M. Segall; Roland Wolkowicz; Christopher T. Cornell; J. Lindsay Whitton; Roberta A. Gottlieb; Ralph Feuer

Coxsackievirus B3 (CVB3), a member of the picornavirus family and enterovirus genus, causes viral myocarditis, aseptic meningitis, and pancreatitis in humans. We genetically engineered a unique molecular marker, “fluorescent timer” protein, within our infectious CVB3 clone and isolated a high-titer recombinant viral stock (Timer-CVB3) following transfection in HeLa cells. “Fluorescent timer” protein undergoes slow conversion of fluorescence from green to red over time, and Timer-CVB3 can be utilized to track virus infection and dissemination in real time. Upon infection with Timer-CVB3, HeLa cells, neural progenitor and stem cells (NPSCs), and C2C12 myoblast cells slowly changed fluorescence from green to red over 72 hours as determined by fluorescence microscopy or flow cytometric analysis. The conversion of “fluorescent timer” protein in HeLa cells infected with Timer-CVB3 could be interrupted by fixation, suggesting that the fluorophore was stabilized by formaldehyde cross-linking reactions. Induction of a type I interferon response or ribavirin treatment reduced the progression of cell-to-cell virus spread in HeLa cells or NPSCs infected with Timer-CVB3. Time lapse photography of partially differentiated NPSCs infected with Timer-CVB3 revealed substantial intracellular membrane remodeling and the assembly of discrete virus replication organelles which changed fluorescence color in an asynchronous fashion within the cell. “Fluorescent timer” protein colocalized closely with viral 3A protein within virus replication organelles. Intriguingly, infection of partially differentiated NPSCs or C2C12 myoblast cells induced the release of abundant extracellular microvesicles (EMVs) containing matured “fluorescent timer” protein and infectious virus representing a novel route of virus dissemination. CVB3 virions were readily observed within purified EMVs by transmission electron microscopy, and infectious virus was identified within low-density isopycnic iodixanol gradient fractions consistent with membrane association. The preferential detection of the lipidated form of LC3 protein (LC3 II) in released EMVs harboring infectious virus suggests that the autophagy pathway plays a crucial role in microvesicle shedding and virus release, similar to a process previously described as autophagosome-mediated exit without lysis (AWOL) observed during poliovirus replication. Through the use of this novel recombinant virus which provides more dynamic information from static fluorescent images, we hope to gain a better understanding of CVB3 tropism, intracellular membrane reorganization, and virus-associated microvesicle dissemination within the host.


Autophagy | 2016

Mitophagy is required for mitochondrial biogenesis and myogenic differentiation of C2C12 myoblasts

Jon Sin; Allen M. Andres; David J. R. Taylor; Thomas A. Weston; Yoshimi Hiraumi; Aleksandr Stotland; Brandon J. Kim; Chengqun Huang; Kelly S. Doran; Roberta A. Gottlieb

ABSTRACT Myogenesis is a crucial process governing skeletal muscle development and homeostasis. Differentiation of primitive myoblasts into mature myotubes requires a metabolic switch to support the increased energetic demand of contractile muscle. Skeletal myoblasts specifically shift from a highly glycolytic state to relying predominantly on oxidative phosphorylation (OXPHOS) upon differentiation. We have found that this phenomenon requires dramatic remodeling of the mitochondrial network involving both mitochondrial clearance and biogenesis. During early myogenic differentiation, autophagy is robustly upregulated and this coincides with DNM1L/DRP1 (dynamin 1-like)-mediated fragmentation and subsequent removal of mitochondria via SQSTM1 (sequestosome 1)-mediated mitophagy. Mitochondria are then repopulated via PPARGC1A/PGC-1α (peroxisome proliferator-activated receptor gamma, coactivator 1 alpha)-mediated biogenesis. Mitochondrial fusion protein OPA1 (optic atrophy 1 [autosomal dominant]) is then briskly upregulated, resulting in the reformation of mitochondrial networks. The final product is a myotube replete with new mitochondria. Respirometry reveals that the constituents of these newly established mitochondrial networks are better primed for OXPHOS and are more tightly coupled than those in myoblasts. Additionally, we have found that suppressing autophagy with various inhibitors during differentiation interferes with myogenic differentiation. Together these data highlight the integral role of autophagy and mitophagy in myogenic differentiation.


Circulation Research | 2015

Untangling Autophagy Measurements: All Fluxed Up

Roberta A. Gottlieb; Allen M. Andres; Jon Sin; David J. Taylor

Autophagy is an important physiological process in the heart, and alterations in autophagic activity can exacerbate or mitigate injury during various pathological processes. Methods to assess autophagy have changed rapidly because the field of research has expanded. As with any new field, methods and standards for data analysis and interpretation evolve as investigators acquire experience and insight. The purpose of this review is to summarize current methods to measure autophagy, selective mitochondrial autophagy (mitophagy), and autophagic flux. We will examine several published studies where confusion arose in data interpretation, to illustrate the challenges. Finally, we will discuss methods to assess autophagy in vivo and in patients.


Autophagy | 2013

MitoTimer: a novel tool for monitoring mitochondrial turnover.

Genaro Hernandez; Christine A Thornton; Aleksandr Stotland; Diana Lui; Jon Sin; Jennifer Ramil; Najib Magee; Allen M. Andres; Giovanni Quarato; Raquel S. Carreira; M. Richard Sayen; Roland Wolkowicz; Roberta A. Gottlieb

Fluorescent Timer, or DsRed1-E5, is a mutant of the red fluorescent protein, dsRed, in which fluorescence shifts over time from green to red as the protein matures. This molecular clock gives temporal and spatial information on protein turnover. To visualize mitochondrial turnover, we targeted Timer to the mitochondrial matrix with a mitochondrial-targeting sequence (coined “MitoTimer”) and cloned it into a tetracycline-inducible promoter construct to regulate its expression. Here we report characterization of this novel fluorescent reporter for mitochondrial dynamics. Tet-On HEK 293 cells were transfected with pTRE-tight-MitoTimer and production was induced with doxycycline (Dox). Mitochondrial distribution was demonstrated by fluorescence microscopy and verified by subcellular fractionation and western blot analysis. Dox addition for as little as 1 h was sufficient to induce MitoTimer expression within 4 h, with persistence in the mitochondrial fraction for up to 6 d. The color-specific conformation of MitoTimer was stable after fixation with 4% paraformaldehyde. Ratiometric analysis of MitoTimer revealed a time-dependent transition from green to red over 48 h and was amenable to analysis by fluorescence microscopy and flow cytometry of whole cells or isolated mitochondria. A second Dox administration 48 h after the initial induction resulted in a second round of expression of green MitoTimer. The extent of new protein incorporation during a second pulse was increased by administration of a mitochondrial uncoupler or simvastatin, both of which trigger mitophagy and biogenesis. MitoTimer is a novel fluorescent reporter protein that can reveal new insights into mitochondrial dynamics within cells. Coupled with organelle flow cytometry, it offers new opportunities to investigate mitochondrial subpopulations by biochemical or proteomic methods.


Virology | 2015

Recent progress in understanding coxsackievirus replication, dissemination, and pathogenesis

Jon Sin; Vrushali Mangale; Wdee Thienphrapa; Roberta A. Gottlieb; Ralph Feuer

Coxsackieviruses (CVs) are relatively common viruses associated with a number of serious human diseases, including myocarditis and meningo-encephalitis. These viruses are considered cytolytic yet can persist for extended periods of time within certain host tissues requiring evasion from the host immune response and a greatly reduced rate of replication. A member of Picornaviridae family, CVs have been historically considered non-enveloped viruses - although recent evidence suggest that CV and other picornaviruses hijack host membranes and acquire an envelope. Acquisition of an envelope might provide distinct benefits to CV virions, such as resistance to neutralizing antibodies and efficient nonlytic viral spread. CV exhibits a unique tropism for progenitor cells in the host which may help to explain the susceptibility of the young host to infection and the establishment of chronic disease in adults. CVs have also been shown to exploit autophagy to maximize viral replication and assist in unconventional release from target cells. In this article, we review recent progress in clarifying virus replication and dissemination within the host cell, identifying determinants of tropism, and defining strategies utilized by the virus to evade the host immune response. Also, we will highlight unanswered questions and provide future perspectives regarding the potential mechanisms of CV pathogenesis.


Methods of Molecular Biology | 2015

Measuring Cardiac Autophagic Flux In Vitro and In Vivo

Michael A. Gurney; Chengqun Huang; Jennifer Ramil; Nandini Ravindran; Allen M. Andres; Jon Sin; Phyllis-Jean Linton; Roberta A. Gottlieb

Autophagy is a lysosomal-dependent catabolic pathway that recycles various cytoplasmic-borne components, such as organelles and proteins, through the lysosomes. This process creates energy and biomolecules that are used to maintain homeostasis and to serve as an energy source under conditions of acute stress. Autophagic flux is a measure of efficiency or throughput of the pathway. Here, we describe a method for determining autophagic flux in vitro and in vivo using the autophagosomal/lysosomal fusion inhibitors chloroquine or bafilomycin A1 and then probing for the autophagosomal marker LC3-II via Western Blot.


Journal of Virology | 2017

Coxsackievirus B Escapes the Infected Cell in Ejected Mitophagosomes

Jon Sin; Laura L. McIntyre; Aleksandr Stotland; Ralph Feuer; Roberta A. Gottlieb

ABSTRACT Coxsackievirus B (CVB) is a common enterovirus that can cause various systemic inflammatory diseases. Because CVB lacks an envelope, it has been thought to be inherently cytolytic, wherein CVB can escape from the infected host cell only by causing it to rupture. In recent years, however, we and others have observed that various naked viruses, such as CVB, can trigger the release of infectious extracellular microvesicles (EMVs) that contain viral material. This mode of cellular escape has been suggested to allow the virus to be masked from the adaptive immune system. Additionally, we have previously reported that these viral EMVs have LC3, suggesting that they originated from autophagosomes. We now report that CVB-infected cells trigger DRP1-mediated fragmentation of mitochondria, which is a precursor to autophagic mitochondrial elimination (mitophagy). However, rather than being degraded by lysosomes, mitochondrion-containing autophagosomes are released from the cell. We believe that CVB localizes to mitochondria, induces mitophagy, and subsequently disseminates from the cell in an autophagosome-bound mitochondrion-virus complex. Suppressing the mitophagy pathway in HL-1 cardiomyocytes with either small interfering RNA (siRNA) or Mdivi-1 caused marked reduction in virus production. The findings in this study suggest that CVB subverts mitophagy machinery to support viral dissemination in released EMVs. IMPORTANCE Coxsackievirus B (CVB) can cause a number of life-threatening inflammatory diseases. Though CVB is well known to disseminate via cytolysis, recent reports have revealed a second pathway in which CVB can become encapsulated in host membrane components to escape the cell in an exosome-like particle. Here we report that these membrane-bound structures derive from mitophagosomes. Blocking various steps in the mitophagy pathway reduced levels of intracellular and extracellular virus. Not only does this study reveal a novel mechanism of picornaviral dissemination, but also it sheds light on new therapeutic targets to treat CVB and potentially other picornaviral infections.


PLOS Pathogens | 2014

The impact of juvenile coxsackievirus infection on cardiac progenitor cells and postnatal heart development.

Jon Sin; Jenna M. Puccini; Chengqun Huang; Mathias Konstandin; Paul E. Gilbert; Mark A. Sussman; Roberta A. Gottlieb; Ralph Feuer

Coxsackievirus B (CVB) is an enterovirus that most commonly causes a self-limited febrile illness in infants, but cases of severe infection can manifest in acute myocarditis. Chronic consequences of mild CVB infection are unknown, though there is an epidemiologic association between early subclinical infections and late heart failure, raising the possibility of subtle damage leading to late-onset dysfunction, or chronic ongoing injury due to inflammatory reactions during latent infection. Here we describe a mouse model of juvenile infection with a subclinical dose of coxsackievirus B3 (CVB3) which showed no evident symptoms, either immediately following infection or in adult mice. However following physiological or pharmacologically-induced cardiac stress, juvenile-infected adult mice underwent cardiac hypertrophy and dilation indicative of progression to heart failure. Evaluation of the vasculature in the hearts of adult mice subjected to cardiac stress showed a compensatory increase in CD31+ blood vessel formation, although this effect was suppressed in juvenile-infected mice. Moreover, CVB3 efficiently infected juvenile c-kit+ cells, and cardiac progenitor cell numbers were reduced in the hearts of juvenile-infected adult mice. These results suggest that the exhausted cardiac progenitor cell pool following juvenile CVB3 infection may impair the hearts ability to increase capillary density to adapt to increased load.


Circulation Research | 2015

Abstract 242: Mitophagy is Required for Mitochondrial Biogenesis and Myogenic Differentiation of Myoblasts

Jon Sin; Allen M. Andres; David J. Taylor; Aleksandr Stotland; Chengqun Huang; Roberta A. Gottlieb


PLOS Pathogens | 2014

Colocalization of “fluorescent timer” protein and viral 3A protein in NPSCs infected with Timer-CVB3.

Scott M. Robinson; Ginger Tsueng; Jon Sin; Vrushali Mangale; Shahad Rahawi; Laura L. McIntyre; Wesley Williams; Nelson Kha; Casey Cruz; Bryan M. Hancock; David P. Nguyen; M. Richard Sayen; Brett J. Hilton; Kelly S. Doran; Anca M. Segall; Roland Wolkowicz; Christopher T. Cornell; J. Lindsay Whitton; Roberta A. Gottlieb; Ralph Feuer

Collaboration


Dive into the Jon Sin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ralph Feuer

San Diego State University

View shared research outputs
Top Co-Authors

Avatar

Allen M. Andres

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Aleksandr Stotland

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Chengqun Huang

San Diego State University

View shared research outputs
Top Co-Authors

Avatar

Laura L. McIntyre

San Diego State University

View shared research outputs
Top Co-Authors

Avatar

Vrushali Mangale

San Diego State University

View shared research outputs
Top Co-Authors

Avatar

Kelly S. Doran

San Diego State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roland Wolkowicz

San Diego State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge