Searched for: school:SOM
Department/Unit:Cell Biology
Altered transcriptome and disease-related phenotype emerge only after fibroblasts harvested from patients with age-related macular degeneration are differentiated into retinal pigment epithelium
Cai, Hui; Gong, Jie; Noggle, Scott; Paull, Daniel; Rizzolo, Lawrence J; Del Priore, Lucian V; Fields, Mark A
We have reported previously that retinal pigment epithelium (RPE) differentiated from induced pluripotent stem cells (iPSC) generated from fibroblasts of patients with age-related macular degeneration (AMD) exhibit a retinal degenerative disease phenotype and a distinct transcriptome compared to age-matched controls. Since the genetic composition of the iPSC and RPE are inherited from fibroblasts, we investigated whether differential behavior was present in the parental fibroblasts and iPSC prior to differentiation of the cell lines into RPE. Principal component analyses revealed significant overlap (essentially no differences) in the transcriptome of fibroblasts between AMD and controls. After reprogramming, there was no significant difference in the transcriptome of iPSC generated from AMD versus normal donors. In contrast, the transcriptome of RPE derived from iPSC segregated into two distinct clusters of AMD-derived cells versus controls. Interestingly, mitochondrial dysfunction in AMD-derived RPE was evident after approximately two months in culture. Moreover, these differences in mitochondrial dysfunction were not evident in the parental fibroblasts and iPSC. This study demonstrates an altered transcriptome and impaired mitochondrial function in RPE derived from AMD patients versus controls, and demonstrates these differences are not present in the original fibroblasts or iPSC. These results suggest that pathology in AMD is triggered upon differentiation of parent cells into RPE. More study of this phenomenon could advance the current understandings of the etiology of AMD and the development of novel therapeutic targets.
PMID: 33895162
ISSN: 1096-0007
CID: 4852852
Design of multi-scale protein complexes by hierarchical building block fusion
Hsia, Yang; Mout, Rubul; Sheffler, William; Edman, Natasha I; Vulovic, Ivan; Park, Young-Jun; Redler, Rachel L; Bick, Matthew J; Bera, Asim K; Courbet, Alexis; Kang, Alex; Brunette, T J; Nattermann, Una; Tsai, Evelyn; Saleem, Ayesha; Chow, Cameron M; Ekiert, Damian; Bhabha, Gira; Veesler, David; Baker, David
A systematic and robust approach to generating complex protein nanomaterials would have broad utility. We develop a hierarchical approach to designing multi-component protein assemblies from two classes of modular building blocks: designed helical repeat proteins (DHRs) and helical bundle oligomers (HBs). We first rigidly fuse DHRs to HBs to generate a large library of oligomeric building blocks. We then generate assemblies with cyclic, dihedral, and point group symmetries from these building blocks using architecture guided rigid helical fusion with new software named WORMS. X-ray crystallography and cryo-electron microscopy characterization show that the hierarchical design approach can accurately generate a wide range of assemblies, including a 43 nm diameter icosahedral nanocage. The computational methods and building block sets described here provide a very general route to de novo designed protein nanomaterials.
PMID: 33863889
ISSN: 2041-1723
CID: 4858772
miR-33 Silencing Reprograms the Immune Cell Landscape in Atherosclerotic Plaques
Afonso, Milessa Silva; Sharma, Monika; Schlegel, Martin Paul; van Solingen, Coen; Koelwyn, Graeme J; Shanley, Lianne C; Beckett, Lauren; Peled, Daniel; Rahman, Karishma; Giannarelli, Chiara; Li, Huilin; Brown, Emily J; Khodadadi-Jamayran, Alireza; Fisher, Edward A; Moore, Kathryn J
Rationale: MicroRNA-33 post-transcriptionally represses genes involved in lipid metabolism and energy homeostasis. Targeted inhibition of miR-33 increases plasma HDL cholesterol and promotes atherosclerosis regression, in part, by enhancing reverse cholesterol transport and dampening plaque inflammation. However, how miR-33 reshapes the immune microenvironment of plaques remains poorly understood. Objective: To define how miR-33 inhibition alters the dynamic balance and transcriptional landscape of immune cells in atherosclerotic plaques. Methods and Results: We used single cell RNA-sequencing of aortic CD45+ cells, combined with immunohistologic, morphometric and flow cytometric analyses to define the changes in plaque immune cell composition, gene expression and function following miR-33 inhibition. We report that anti-miR-33 treatment of Ldlr-/- mice with advanced atherosclerosis reduced plaque burden and altered the plaque immune cell landscape by shifting the balance of pro- and anti-atherosclerotic macrophage and T cell subsets. By quantifying the kinetic processes that determine plaque macrophage burden, we found that anti-miR-33 reduced levels of circulating monocytes and splenic myeloid progenitors, decreased macrophage proliferation and retention, and promoted macrophage attrition by apoptosis and efferocytotic clearance. scRNA-sequencing of aortic arch plaques showed that anti-miR-33 reduced the frequency of MHCIIhi "inflammatory" and Trem2hi "metabolic" macrophages, but not tissue resident macrophages. Furthermore, anti-miR-33 led to derepression of distinct miR-33 target genes in the different macrophage subsets: in resident and Trem2hi macrophages, anti-miR-33 relieved repression of miR-33 target genes involved in lipid metabolism (e.g., Abca1, Ncoa1, Ncoa2, Crot), whereas in MHCIIhi macrophages, anti-miR-33 upregulated target genes involved in chromatin remodeling and transcriptional regulation. Anti-miR-33 also reduced the accumulation of aortic CD8+ T cells and CD4+ Th1 cells, and increased levels of FoxP3+ regulatory T cells in plaques, consistent with an immune-dampening effect on plaque inflammation. Conclusions: Our results provide insight into the immune mechanisms and cellular players that execute anti-miR-33's atheroprotective actions in the plaque.
PMID: 33593073
ISSN: 1524-4571
CID: 4786732
One-pot synthesis of linear triblock terpolymers and their aqueous self-Assembly
Ahmed, Eman; Womble, C. Tyler; Cho, Jinwon; Dancel-Manning, Kristen; Rice, William J.; Jang, Seung Soon; Weck, Marcus
Compartmentalized micelles are prepared through the self-Assembly of linear triblock terpolymers containing hydrophilic (H), lipophilic (L), and fluorophilic (F) domains. The triblock copolymers were synthesized via living ring-opening metathesis polymerization (ROMP) of norbornene-based monomers. Our terpolymer design offers a facile approach for the synthesis of the target materials with fast polymerization kinetics, complete block incorporation and control over block sequence. Various triblock terpolymers are prepared with variations in block sequence and block ratio and self-Assembled in aqueous media. Interaction parameter (χ) values between each block are determined using a Flory-Huggins based computational model. "Core-shell-corona", "disk-like", "raspberry-like"and "worm-like"morphologies are observed through cryogenic transmission electron microscopy and dissipative particle dynamics simulations. This journal is
SCOPUS:85103833988
ISSN: 1759-9954
CID: 4860932
Dominant role of CDKN2B/p15INK4B of 9p21.3 tumor suppressor hub in inhibition of cell-cycle and glycolysis
Xia, Yong; Liu, Yan; Yang, Chao; Simeone, Diane M; Sun, Tung-Tien; DeGraff, David J; Tang, Moon-Shong; Zhang, Yingkai; Wu, Xue-Ru
Human chromosome 9p21.3 is susceptible to inactivation in cell immortalization and diseases, such as cancer, coronary artery disease and type-2 diabetes. Although this locus encodes three cyclin-dependent kinase (CDK) inhibitors (p15INK4B, p14ARF and p16INK4A), our understanding of their functions and modes of action is limited to the latter two. Here, we show that in vitro p15INK4B is markedly stronger than p16INK4A in inhibiting pRb1 phosphorylation, E2F activity and cell-cycle progression. In mice, urothelial cells expressing oncogenic HRas and lacking p15INK4B, but not those expressing HRas and lacking p16INK4A, develop early-onset bladder tumors. The potency of CDKN2B/p15INK4B in tumor suppression relies on its strong binding via key N-terminal residues to and inhibition of CDK4/CDK6. p15INK4B also binds and inhibits enolase-1, a glycolytic enzyme upregulated in most cancer types. Our results highlight the dual inhibition of p15INK4B on cell proliferation, and unveil mechanisms whereby p15INK4B aberrations may underpin cancer and non-cancer conditions.
PMID: 33824349
ISSN: 2041-1723
CID: 4840932
Neuraminidase B controls neuraminidase A-dependent mucus production and evasion
Hammond, Alexandria J; Binsker, Ulrike; Aggarwal, Surya D; Ortigoza, Mila Brum; Loomis, Cynthia; Weiser, Jeffrey N
Binding of Streptococcus pneumoniae (Spn) to nasal mucus leads to entrapment and clearance via mucociliary activity during colonization. To identify Spn factors allowing for evasion of mucus binding, we used a solid-phase adherence assay with immobilized mucus of human and murine origin. Spn bound large mucus particles through interactions with carbohydrate moieties. Mutants lacking neuraminidase A (nanA) or neuraminidase B (nanB) showed increased mucus binding that correlated with diminished removal of terminal sialic acid residues on bound mucus. The non-additive activity of the two enzymes raised the question why Spn expresses two neuraminidases and suggested they function in the same pathway. Transcriptional analysis demonstrated expression of nanA depends on the enzymatic function of NanB. As transcription of nanA is increased in the presence of sialic acid, our findings suggest that sialic acid liberated from host glycoconjugates by the secreted enzyme NanB induces the expression of the cell-associated enzyme NanA. The absence of detectable mucus desialylation in the nanA mutant, in which NanB is still expressed, suggests that NanA is responsible for the bulk of the modification of host glycoconjugates. Thus, our studies describe a functional role for NanB in sialic acid sensing in the host. The contribution of the neuraminidases in vivo was then assessed in a murine model of colonization. Although mucus-binding mutants showed an early advantage, this was only observed in a competitive infection, suggesting a complex role of neuraminidases. Histologic examination of the upper respiratory tract demonstrated that Spn stimulates mucus production in a neuraminidase-dependent manner. Thus, an increase production of mucus containing secretions appears to be balanced, in vivo, by decreased mucus binding. We postulate that through the combined activity of its neuraminidases, Spn evades mucus binding and mucociliary clearance, which is needed to counter neuraminidase-mediated stimulation of mucus secretions.
PMID: 33819312
ISSN: 1553-7374
CID: 4838982
Breaking Tradition to Bridge Bench and Bedside: Accelerating the MD-PhD-Residency Pathway
Modrek, Aram S; Tanese, Naoko; Placantonakis, Dimitris G; Sulman, Erik P; Rivera, Rafael; Du, Kevin L; Gerber, Naamit K; David, Gregory; Chesler, Mitchell; Philips, Mark R; Cangiarella, Joan
PROBLEM/OBJECTIVE:Physician-scientists are individuals trained in both clinical practice and scientific research. Often, the goal of physician-scientist training is to address pressing questions in biomedical research. The established pathways to formally train such individuals are, mainly, MD-PhD programs and physician-scientist track residencies. Although graduates of these pathways are well equipped to be physician-scientists, numerous factors, including funding and length of training, discourage application to such programs and impede success rates. APPROACH/METHODS:To address some of the pressing challenges in training and retaining burgeoning physician-scientists, New York University Grossman School of Medicine formed the Accelerated MD-PhD-Residency Pathway in 2016. This pathway builds on the previously established accelerated three-year MD pathway to residency at the same institution. The Accelerated MD-PhD-Residency Pathway conditionally accepts MD-PhD trainees to a residency position at the same institution through the National Resident Matching Program. OUTCOMES/RESULTS:Since its inception, 2 students have joined the Accelerated MD-PhD-Residency Pathway, which provides protected research time in their chosen residency. The pathway reduces the time to earn an MD and PhD by one year and reduces the MD training phase to three years, reducing the cost and lowering socioeconomic barriers. Remaining at the same institution for residency allows for the growth of strong research collaborations and mentoring opportunities, which foster success. NEXT STEPS/UNASSIGNED:The authors and institutional leaders plan to increase the number of trainees that are accepted into the Accelerated MD-PhD-Residency Pathway and track the success of these students through residency and into practice to determine if the pathway is meeting its goal of increasing the number of practicing physician-scientists. The authors hope this model can serve as an example to leaders at other institutions who may wish to adopt this pathway for the training of their MD-PhD students.
PMID: 33464738
ISSN: 1938-808x
CID: 4760452
A Global eDelphi Exercise to Identify Core Domains and Domain Items for the Development of a Global Registry of Alopecia Areata Disease Severity and Treatment Safety (GRASS)
Wall, Dmitri; Meah, Nekma; York, Katherine; Bhoyrul, Bevin; Bokhari, Laita; Abraham, Leonardo Spagnol; Adams, RoisÃn; Bergfeld, Wilma; Betz, Regina C; Blume-Peytavi, Ulrike; Callender, Valerie; Campbell, Chel; Chambers, Jen; Chen, Gang; Chitreddy, Vijaya; Cotsarelis, George; Craiglow, Brittany; Dhurat, Rachita; Dlova, Ncoza; Donovan, Jeff; Duque-Estrada, Bruna; Eisman, Samantha; Ellison, Abby; Farrant, Paul; Barberá, Juan Ferrando; Gadzhigoroeva, Aida; Grimalt, Ramon; Harries, Matthew; Hordinsky, Maria; Irvine, Alan D; Jolliffe, Victoria; Jones, Leslie; King, Brett; Lee, Won-Soo; Lortkipanidze, Nino; McMichael, Amy; Messenger, Andrew; Mirmirani, Paradi; Olsen, Elise; Orlow, Seth J; Ovcharenko, Yuliya; Piraccini, Bianca Maria; Pirmez, Rodrigo; Rakowska, Adriana; Reygagne, Pascal; Riley, Melissa; Rudnicka, Lidia; Saceda Corralo, David; Shapiro, Jerry; Sharma, Pooja; Silyuk, Tatiana; Kaiumov, Spartak; Tobin, Desmond J; Tosti, Antonella; Vañó-Galván, Sergio; Vogt, Annika; Wade, Martin; Yip, Leona; Zlotogorski, Abraham; Zhou, Cheng; Sinclair, Rodney
Importance/UNASSIGNED:A recent expert consensus exercise emphasized the importance of developing a global network of patient registries for alopecia areata to redress the paucity of comparable, real-world data regarding the effectiveness and safety of existing and emerging therapies for alopecia areata. Objective/UNASSIGNED:To generate core domains and domain items for a global network of alopecia areata patient registries. Evidence Review/UNASSIGNED:Sixty-six participants, representing physicians, patient organizations, scientists, the pharmaceutical industry, and pharmacoeconomic experts, participated in a 3-round eDelphi process, culminating in a face-to-face meeting at the World Congress of Dermatology, Milan, Italy, June 14, 2019. Findings/UNASSIGNED:Ninety-two core data items, across 25 domains, achieved consensus agreement. Twenty further noncore items were retained to facilitate data harmonization in centers that wish to record them. Broad representation across multiple stakeholder groups was sought; however, the opinion of physicians was overrepresented. Conclusions and Relevance/UNASSIGNED:This study identifies the domains and domain items required to develop a global network of alopecia areata registries. These domains will facilitate a standardized approach that will enable the recording of a comprehensive, comparable data set required to oversee the introduction of new therapies and harness real-world evidence from existing therapies at a time when the alopecia areata treatment paradigm is being radically and positively disrupted. Reuse of similar, existing frameworks in atopic dermatitis, produced by the Treatment of Atopic Eczema (TREAT) Registry Taskforce, increases the potential to reuse existing resources, creates opportunities for comparison of data across dermatology subspecialty disease areas, and supports the concept of data harmonization.
PMID: 33656556
ISSN: 2168-6084
CID: 4801512
Cx43 hemichannel microdomain signaling at the intercalated disc enhances cardiac excitability
De Smet, Maarten Aj; Lissoni, Alessio; Nezlobinsky, Timur; Wang, Nan; Dries, Eef; Pérez-Hernández, Marta; Lin, Xianming; Amoni, Matthew; Vervliet, Tim; Witschas, Katja; Rothenberg, Eli; Bultynck, Geert; Schulz, Rainer; Panfilov, Alexander V; Delmar, Mario; Sipido, Karin R; Leybaert, Luc
Cx43, a major cardiac connexin, forms precursor hemichannels that accrue at the intercalated disc to assemble as gap junctions. While gap junctions are crucial for electrical conduction in the heart, little is known on potential roles of hemichannels. Recent evidence suggests that inhibiting Cx43 hemichannel opening with Gap19 has antiarrhythmic effects. Here, we used multiple electrophysiology, imaging and super-resolution techniques to understand and define the conditions underlying Cx43 hemichannel activation in ventricular cardiomyocytes, their contribution to diastolic Ca2+ release from the sarcoplasmic reticulum, and their impact on electrical stability. We showed that Cx43 hemichannels are activated during diastolic Ca2+ release in single ventricular cardiomyocytes and cardiomyocyte cell pairs from mouse and pig. This activation involved Cx43 hemichannel Ca2+ entry and coupling to Ca2+ release microdomains at the intercalated disc resulting in enhanced Ca2+ dynamics. Hemichannel opening furthermore contributed to delayed afterdepolarizations and triggered action potentials. In single cardiomyocytes, cardiomyocyte cell pairs and arterially perfused tissue wedges from failing human hearts, increased hemichannel activity contributed to electrical instability as compared to non-failing rejected donor hearts. We conclude that microdomain coupling between Cx43 hemichannels and Ca2+ release is a novel, targetable, mechanism of cardiac arrhythmogenesis in heart failure.
PMID: 33621213
ISSN: 1558-8238
CID: 4794482
Progranulin promotes bone fracture healing via TNFR pathways in mice with type 2 diabetes mellitus
Ding, Yuanjing; Wei, Jianlu; Hettinghouse, Aubryanna; Li, Guangfei; Li, Xin; Einhorn, Thomas A; Liu, Chuan-Ju
Type 2 diabetes mellitus (T2DM) significantly increases bone fragility and fracture risk. Progranulin (PGRN) promotes bone fracture healing in both physiological and type 1 diabetic conditions. The present study aimed to investigate the role of PGRN in T2DM bone fracture healing. MKR mice (with an FVB/N genetic background) were used as the T2DM model. Drill-hole and Bonnarens and Einhorn models were used to investigate the role of PGRN in T2DM fracture healing in vivo. Primary bone marrow cells were isolated for molecular and signaling studies, and reverse transcription-polymerase chain reaction, immunohistochemical staining, and western blotting were performed to assess PGRN effects in vitro. PGRN mRNA and protein expression were upregulated in the T2DM model. Local administration of recombinant PGRN effectively promoted T2DM bone fracture healing in vivo. Additionally, PGRN could induce anabolic metabolism during endochondral ossification through the TNFR2-Akt and Erk1/2 pathways. Furthermore, PGRN showed anti-inflammatory activity in the T2DM bone regeneration process. These findings suggest that local administration of exogenous PGRN may be an alternative strategy to support bone regeneration in patients with T2DM. Additionally, PGRN might hold therapeutic potential for other TNFR-related metabolic disorders.
PMID: 33543485
ISSN: 1749-6632
CID: 4776682