Try a new search

Format these results:

Searched for:

school:SOM

Department/Unit:Cell Biology

Total Results:

14089


Penfluridol targets acid sphingomyelinase to inhibit TNF signaling and is therapeutic against inflammatory autoimmune diseases

Chen, Yue-Hong; Liu, Rong-Han; Cui, Ya-Zhou; Hettinghouse, Aubryanna; Fu, Wen-Yu; Zhang, Lei; Zhang, Chen; Liu, Chuan-Ju
BACKGROUND:Penfluridol, isolated from an FDA-approved small-molecule drug library as an inhibitor of tumor necrosis factor α (TNFα)-stimulated NF-κB activation, is clinically used to treat chronic schizophrenia and related disorders. This study is aimed to investigate the therapeutic effect of penfluridol on TNFα-stimulated inflammatory autoimmune diseases, particularly inflammatory arthritis. METHODS:Various in vitro studies to confirm the inhibitory effect of penfluridol on TNFα-induced NF-κB activity in bone marrow-derived macrophages or Raw 264.7 macrophage cell line. In vivo studies assessed the therapeutic effects of penfluridol in various disease models, including TNFα transgenic mice, collagen-induced arthritis, DSS-induced colitis, and TNBS-induced colitis. Identification and characterization of the binding of penfluridol to acid sphingomyelinase using bioinformatics and drug affinity responsive target stability assay. Acid sphingomyelinase activity assays to reveal penfluridol-mediated inhibition of acid sphingomyelinase activity. siRNA knockdown experiments to illustrate the dependence of penfluridol's anti-TNF activity on acid sphingomyelinase. RESULTS:Penfluridol effectively inhibited TNFα-induced NF-κB activation in vitro and alleviated the severity of arthritis and colitis in vivo. Mechanistic studies revealed that penfluridol bound to acid sphingomyelinase and inhibited its activation. In addition, knockdown of acid sphingomyelinase largely abolished the inhibitory effects of penfluridol on TNFα-induced inflammatory cytokine production. Furthermore, penfluridol suppressed the differentiation of spleen naive CD4+T cells to TH1 and TH17 and inhibited M1 macrophage polarization. CONCLUSION/CONCLUSIONS:This study provides the rationale for the possible innovative use of penfluridol as a newly identified small-molecule drug for TNFα-driven diseases, such as inflammatory arthritis and colitis.
PMCID:8767691
PMID: 35045889
ISSN: 1478-6362
CID: 5131602

Injectable recombinant block polymer gel for sustained delivery of therapeutic protein in post traumatic osteoarthritis

Katyal, Priya; Hettinghouse, Aubryanna; Meleties, Michael; Hasan, Sadaf; Chen, Changhong; Cui, Min; Sun, Guodong; Menon, Rajiv; Lin, Bonnie; Regatte, Ravinder; Montclare, Jin Kim; Liu, Chuan-Ju
Protein-based biomaterials offer several advantages over synthetic materials, owing to their unique stimuli-responsive properties, biocompatibility and modular nature. Here, we demonstrate that E5C, a recombinant protein block polymer, consisting of five repeats of elastin like polypeptide (E) and a coiled-coil domain of cartilage oligomeric matrix protein (C), is capable of forming a porous networked gel at physiological temperature, making it an excellent candidate for injectable biomaterials. Combination of E5C with Atsttrin, a chondroprotective engineered derivative of anti-inflammatory growth factor progranulin, provides a unique biochemical and biomechanical environment to protect against post-traumatic osteoarthritis (PTOA) onset and progression. E5C gel was demonstrated to provide prolonged release of Atsttrin and inhibit chondrocyte catabolism while facilitating anabolic signaling in vitro. We also provide in vivo evidence that prophylactic and therapeutic application of Atsttrin-loaded E5C gels protected against PTOA onset and progression in a rabbit anterior cruciate ligament transection model. Collectively, we have developed a unique protein-based gel capable of minimally invasive, sustained delivery of prospective therapeutics, particularly the progranulin-derivative Atsttrin, for therapeutic application in OA.
PMID: 35032910
ISSN: 1878-5905
CID: 5119242

The transcription factor ZIP-1 promotes resistance to intracellular infection in Caenorhabditis elegans

Lažetić, Vladimir; Wu, Fengting; Cohen, Lianne B; Reddy, Kirthi C; Chang, Ya-Ting; Gang, Spencer S; Bhabha, Gira; Troemel, Emily R
Defense against intracellular infection has been extensively studied in vertebrate hosts, but less is known about invertebrate hosts; specifically, the transcription factors that induce defense against intracellular intestinal infection in the model nematode Caenorhabditis elegans remain understudied. Two different types of intracellular pathogens that naturally infect the C. elegans intestine are the Orsay virus, which is an RNA virus, and microsporidia, which comprise a phylum of fungal pathogens. Despite their molecular differences, these pathogens induce a common host transcriptional response called the intracellular pathogen response (IPR). Here we show that zip-1 is an IPR regulator that functions downstream of all known IPR-activating and regulatory pathways. zip-1 encodes a putative bZIP transcription factor, and we show that zip-1 controls induction of a subset of genes upon IPR activation. ZIP-1 protein is expressed in the nuclei of intestinal cells, and is at least partially required in the intestine to upregulate IPR gene expression. Importantly, zip-1 promotes resistance to infection by the Orsay virus and by microsporidia in intestinal cells. Altogether, our results indicate that zip-1 represents a central hub for triggers of the IPR, and that this transcription factor has a protective function against intracellular pathogen infection in C. elegans.
PMID: 35013162
ISSN: 2041-1723
CID: 5118522

Deficiency of inactive rhomboid protein 2 (iRhom2) attenuates diet-induced hyperlipidemia and early atherogenesis

Hannemann, Carmen; Schecker, Johannes H; Brettschneider, Alica; Grune, Jana; Rösener, Nicole; Weller, Andrea; Stangl, Verena; Fisher, Edward A; Stangl, Karl; Ludwig, Antje; Hewing, Bernd
AIMS/OBJECTIVE:Atherosclerosis is a chronic inflammatory disease of the arterial vessel wall and anti-inflammatory treatment strategies are currently pursued to lower cardiovascular disease burden. Modulation of recently discovered inactive rhomboid protein 2 (iRhom2) attenuates shedding of tumor necrosis factor-alpha (TNF-α) selectively from immune cells. The present study aims at investigating the impact of iRhom2 deficiency on the development of atherosclerosis. METHODS AND RESULTS/RESULTS:Low-density lipoprotein receptor (LDLR)-deficient mice with additional deficiency of iRhom2 (LDLR-/-iRhom2-/-) and control (LDLR-/-) mice were fed a Western type diet (WD) for 8 or 20 weeks to induce early or advanced atherosclerosis. Deficiency of iRhom2 resulted in a significant decrease in the size of early atherosclerotic plaques as determined in aortic root cross sections. LDLR-/-iRhom2-/- mice exhibited significantly lower serum levels of TNF-α and lower circulating and hepatic levels of cholesterol and triglycerides compared to LDLR-/- mice at 8 weeks of WD. Analyses of hepatic bile acid concentration and gene expression at 8 weeks of WD revealed that iRhom2 deficiency prevented WD-induced repression of hepatic bile acid synthesis in LDLR-/- mice. In contrast, at 20 weeks of WD plaque size, plaque composition, and serum levels of TNF-α or cholesterol were not different between genotypes. CONCLUSIONS:Modulation of inflammation by iRhom2 deficiency attenuated diet induced hyperlipidemia and early atherogenesis in LDLR-/- mice. iRhom2 deficiency did not affect diet- induced plaque burden and composition in advanced atherosclerosis in LDLR-/- mice. TRANSLATIONAL PERSPECTIVE/UNASSIGNED:iRhom2 attenuates shedding of TNF-α selectively from immune cells and therefore has emerged as a potential new target for the treatment of inflammatory diseases. In the present study, we identified iRhom2 as a critical link between inflammation, lipid metabolism, and atherogenesis. Selective iRhom2 inhibition represents a potential treatment strategy to modify atherosclerosis, particularly in the presence of enhanced inflammation as observed with diabetes mellitus or rheumatoid arthritis.
PMID: 33576385
ISSN: 1755-3245
CID: 4806462

Modulating the systemic and local adaptive immune response after fracture improves bone regeneration during aging

Lopez, Emma Muiños; Leclerc, Kevin; Ramsukh, Malissa; Parente, Paulo El; Patel, Karan; Aranda, Carlos J; Josephson, Anna M; Remark, Lindsey H; Kirby, David J; Buchalter, Daniel B; Hadi, Tarik; Morgani, Sophie M; Ramkhelawon, Bhama; Leucht, Philipp
Tissue injury leads to the well-orchestrated mobilization of systemic and local innate and adaptive immune cells. During aging, immune cell recruitment is dysregulated, resulting in an aberrant inflammatory response that is detrimental for successful healing. Here, we precisely define the systemic and local immune cell response after femur fracture in young and aging mice and identify increased toll-like receptor signaling as a potential culprit for the abnormal immune cell recruitment observed in aging animals. Myd88, an upstream regulator of TLR-signaling lies at the core of this aging phenotype, and local treatment of femur fractures with a Myd88 antagonist in middle-aged mice reverses the aging phenotype of impaired fracture healing, thus offering a promising therapeutic target that could overcome the negative impact of aging on bone regeneration.
PMID: 34998981
ISSN: 1873-2763
CID: 5118182

Analysis of the Biomarkers for Neurodegenerative Diseases in Aged Progranulin Deficient Mice

Zhao, Xiangli; Hasan, Sadaf; Liou, Benjamin; Lin, Yi; Sun, Ying; Liu, Chuanju
Neurodegenerative diseases are debilitating impairments that affect millions of people worldwide and are characterized by progressive degeneration of structure and function of the central or peripheral nervous system. Effective biomarkers for neurodegenerative diseases can be used to improve the diagnostic workup in the clinic as well as facilitate the development of effective disease-modifying therapies. Progranulin (PGRN) has been reported to be involved in various neurodegenerative disorders. Hence, in the current study we systematically compared the inflammation and accumulation of typical neurodegenerative disease markers in the brain tissue between PGRN knockout (PGRN KO) and wildtype (WT) mice. We found that PGRN deficiency led to significant neuron loss as well as activation of microglia and astrocytes in aged mice. Several characteristic neurodegenerative markers, including α-synuclein, TAR DNA-binding protein 43 (TDP-43), Tau, and β-amyloid, were all accumulated in the brain of PGRN-deficient mice as compared to WT mice. Moreover, higher aggregation of lipofuscin was observed in the brain tissue of PGRN-deficient mice compared with WT mice. In addition, the autophagy was also defective in the brain of PGRN-deficient mice, indicated by the abnormal expression level of autophagy marker LC3-II. Collectively, comprehensive assays support the idea that PGRN plays an important role during the development of neurodegenerative disease, indicating that PGRN might be a useful biomarker for neurodegenerative diseases in clinical settings.
PMID: 35054815
ISSN: 1422-0067
CID: 5131762

ETHANOL-NUTRIENT INTERACTION AND THE FETUS

Chapter by: Fisher, Stanley E.; Karl, Peter I.
in: Fetal Alcohol Syndrome: From Mechanism to Prevention by
[S.l.] : CRC Press, 2022
pp. 269-283
ISBN: 9780849376856
CID: 5349892

Genome-wide association study and functional validation implicates JADE1 in tauopathy

Farrell, Kurt; Kim, SoongHo; Han, Natalia; Iida, Megan A; Gonzalez, Elias M; Otero-Garcia, Marcos; Walker, Jamie M; Richardson, Timothy E; Renton, Alan E; Andrews, Shea J; Fulton-Howard, Brian; Humphrey, Jack; Vialle, Ricardo A; Bowles, Kathryn R; de Paiva Lopes, Katia; Whitney, Kristen; Dangoor, Diana K; Walsh, Hadley; Marcora, Edoardo; Hefti, Marco M; Casella, Alicia; Sissoko, Cheick T; Kapoor, Manav; Novikova, Gloriia; Udine, Evan; Wong, Garrett; Tang, Weijing; Bhangale, Tushar; Hunkapiller, Julie; Ayalon, Gai; Graham, Robert R; Cherry, Jonathan D; Cortes, Etty P; Borukov, Valeriy Y; McKee, Ann C; Stein, Thor D; Vonsattel, Jean-Paul; Teich, Andy F; Gearing, Marla; Glass, Jonathan; Troncoso, Juan C; Frosch, Matthew P; Hyman, Bradley T; Dickson, Dennis W; Murray, Melissa E; Attems, Johannes; Flanagan, Margaret E; Mao, Qinwen; Mesulam, M-Marsel; Weintraub, Sandra; Woltjer, Randy L; Pham, Thao; Kofler, Julia; Schneider, Julie A; Yu, Lei; Purohit, Dushyant P; Haroutunian, Vahram; Hof, Patrick R; Gandy, Sam; Sano, Mary; Beach, Thomas G; Poon, Wayne; Kawas, Claudia H; Corrada, María M; Rissman, Robert A; Metcalf, Jeff; Shuldberg, Sara; Salehi, Bahar; Nelson, Peter T; Trojanowski, John Q; Lee, Edward B; Wolk, David A; McMillan, Corey T; Keene, C Dirk; Latimer, Caitlin S; Montine, Thomas J; Kovacs, Gabor G; Lutz, Mirjam I; Fischer, Peter; Perrin, Richard J; Cairns, Nigel J; Franklin, Erin E; Cohen, Herbert T; Raj, Towfique; Cobos, Inma; Frost, Bess; Goate, Alison; White Iii, Charles L; Crary, John F
Primary age-related tauopathy (PART) is a neurodegenerative pathology with features distinct from but also overlapping with Alzheimer disease (AD). While both exhibit Alzheimer-type temporal lobe neurofibrillary degeneration alongside amnestic cognitive impairment, PART develops independently of amyloid-β (Aβ) plaques. The pathogenesis of PART is not known, but evidence suggests an association with genes that promote tau pathology and others that protect from Aβ toxicity. Here, we performed a genetic association study in an autopsy cohort of individuals with PART (n = 647) using Braak neurofibrillary tangle stage as a quantitative trait. We found some significant associations with candidate loci associated with AD (SLC24A4, MS4A6A, HS3ST1) and progressive supranuclear palsy (MAPT and EIF2AK3). Genome-wide association analysis revealed a novel significant association with a single nucleotide polymorphism on chromosome 4 (rs56405341) in a locus containing three genes, including JADE1 which was significantly upregulated in tangle-bearing neurons by single-soma RNA-seq. Immunohistochemical studies using antisera targeting JADE1 protein revealed localization within tau aggregates in autopsy brains with four microtubule-binding domain repeats (4R) isoforms and mixed 3R/4R, but not with 3R exclusively. Co-immunoprecipitation in post-mortem human PART brain tissue revealed a specific binding of JADE1 protein to four repeat tau lacking N-terminal inserts (0N4R). Finally, knockdown of the Drosophila JADE1 homolog rhinoceros (rno) enhanced tau-induced toxicity and apoptosis in vivo in a humanized 0N4R mutant tau knock-in model, as quantified by rough eye phenotype and terminal deoxynucleotidyl transferase dUTP nick end-labeling (TUNEL) in the fly brain. Together, these findings indicate that PART has a genetic architecture that partially overlaps with AD and other tauopathies and suggests a novel role for JADE1 as a modifier of neurofibrillary degeneration.
PMID: 34719765
ISSN: 1432-0533
CID: 5095122

Exploiting Canonical TGFβ Signaling in Cancer Treatment

Liu, Qi; Chen, Genwen; Moore, Jade; Guix, Ines; Placantonokis, Dimitris; Barcellos-Hoff, Mary Helen
Transforming growth factor β (TGFβ) is a pleiotropic cytokine that plays critical roles to define cancer cell phenotypes, construct the tumor microenvironment, and suppress anti-tumor immune responses. As such, TGFβ is a lynchpin for integrating cancer cell intrinsic pathways and communication among host cells in the tumor and beyond that together affect responses to genotoxic, targeted, and immune therapy. Despite decades of preclinical and clinical studies, evidence of clinical benefit from targeting TGFβ in cancer remains elusive. Here, we review the mechanisms by which TGFβ acts to oppose successful cancer therapy, the reported prognostic and predictive value of TGFβ biomarkers, and the potential impact of inhibiting TGFβ in precision oncology. Paradoxically, the diverse mechanisms by which TGFβ impedes therapeutic response are a principal barrier to implementing TGFβ inhibitors because it is unclear which TGFβ mechanism is functional in which patient. Companion diagnostic tools and specific biomarkers of TGFβ targeted biology will be the key to exploiting TGFβ biology for patient benefit.
PMID: 34670783
ISSN: 1538-8514
CID: 5043352

Simultaneous checkpoint inhibition and immune cell activation that is safely localized to solidtumors [Meeting Abstract]

Richieri, R A; Narula, N; Loomis, C A; Mezzano, V; Billimek, J; Reynolds, G T; Reutelingsperger, C; Zijlstra, A; Parseghian, M H
Unlike other checkpoint inhibitors, our targeted immunotherapeutic localizes to any solid tumor and simultaneouslyshields an agent of immuno suppression while presenting a signal for immunostimulation. Phosphatidylserine (PS)exposure on the extracellular surface of living tumor cells and their vasculatures provides one avenue by which thetumor microenvironment promotes immunosuppression. Extracellular surface PS is inherent to a tumor and itsvasculature, even for inoperable tumors, and its expression cannot be mutated nor affected by acquired drugresistance. Annexin A5 (AnxA5) is a direct, high-affinity PS-binding protein that localizes to cells with PS exposed onthe outer plasma membrane. In our studies, we conjugated a proprietary modified AnxA5, lacking cellularinternalization, to TNFalpha (AnxA5 -TNFalpha) to convert the immunosuppresive environs of a murine 4T1 triplenegative breast cancer (TNBC) into an immunostimulated one. This strategy localized the immune response to the tumor and minimized side effects, as evidenced by a lack of toxicity for up to 7 days in non-tumor bearing Balb/cfemale mice given up to 1 mg/kg. Proper assembly and functionality of AnxA5 -TNFalpha was verified simultaneouslyby ellipsometry, an optical technique similar to plasmon resonance. Fully assembled constructs were tested forbinding to PS coated slides. The degree of light polarization is proportional to the amount of PS bound by the AnxA5complex. Samples could be further incubated with TNF receptors to verify TNFalpha activity. Based on dose escalationstudies in 4T1 tumor-bearing mice where the TNBC tumors were grown in the mammary fat pads, optimal dosages were determined for AnxA5 -TNFalpha (18 mug) and AnxA5 alone as a control (180 mug). These doses were furthertested in a 4T1 growth inhibition study. Tumor size was tracked by caliper in two groups of mice (n=5/group)receiving drug treatment on days 12, 14 and 16 and a repeated measures ANOVA was conducted onmeasurements taken before, during and post-treatment. While median tumor size did not differ between control and drug treatment groups during the pre-treatment interval (p=0.84), there was a significant difference post-treatment(p<0.001) with mice receiving AnxA5 -TNFalpha having much smaller TNBC tumors. Tumors from the study were embedded in paraffin, sectioned (5 mum) and the overall immune cell content determined by H&E staining. Once it was evident there was a greater quantity of immune cells in AnxA5 -TNFalpha treated tumors vs. controls, sections were stained with validated antibodies to identify and count the immunoactivated T-cells, NK-cells and macrophages. There was a 3X greater mean percentage of CD8 and CD4 T-cells in mice receiving drug vs. control(p=0.03) along with 2.5X and 5X increases in NK-cells and M1 immunoactive macrophages, respectively.
Conclusion(s): Our AnxA5 -TNFalpha inhibits the PS inhibitor while simultaneously activating TNF activators!
EMBASE:637180376
ISSN: 2326-6074
CID: 5158452