1
|
Bajaj P, Brennan RJ, Laurent S, Sauzeat S, Dufault M, Richards B, Adkins K. Transcriptomic analysis in liver spheroids identifies a dog-specific mechanism of hepatotoxicity for amcenestrant. Toxicol Sci 2025; 204:228-241. [PMID: 39886943 DOI: 10.1093/toxsci/kfaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025] Open
Abstract
Therapeutic drugs can sometimes cause adverse effects in a nonclinical species that do not translate to other species, including human. Species-specific (rat, dog, and human) in vitro liver spheroids were employed to understand the human relevance of cholestatic liver injury observed with a selective estrogen receptor degrader (amcenestrant) in dog, but not in rat, during preclinical development. Amcenestrant showed comparable cytotoxicity in liver spheroids from all 3 species; however, its M5 metabolite (RA15400562) showed dog preferential cytotoxicity after 7 days of treatment. Whole genome transcript profiles generated from liver spheroids revealed downregulation of genes related to bile acid synthesis and transport indicative of strong farnesoid X receptor (FXR) antagonism following treatment with both amcenestrant and its M5 metabolite in the dog but not in rat or human. In human spheroids, upregulation of genes for detoxification enzymes indicative of pregnane X receptor (PXR) agonism was observed following amcenestrant treatment, whereas in the dog these genes were downregulated. The M5 metabolite showed gene dysregulation indicating PXR agonism in both rat and human, and antagonism in dog. Analysis of liver samples from a 3-mo dog toxicity study conducted with amcenestrant showed downregulation of several genes associated with PXR and FXR, corroborating the in vitro results. These results support the hypothesis that dogs are uniquely susceptible to cholestatic hepatotoxicity following administration of amcenestrant due to species-specific antagonism of FXR and highlight the value of in vitro liver spheroids to investigating mechanisms of toxicity and possible species differences.
Collapse
Affiliation(s)
- Piyush Bajaj
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02141, United States
| | - Richard J Brennan
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02141, United States
| | | | | | - Michael Dufault
- Precision Medicine and Computational Biology, Sanofi, Cambridge, MA 02141, United States
| | - Brenda Richards
- Genetic Medicine Unit, Sanofi, Waltham, MA 02451, United States
| | - Karissa Adkins
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02141, United States
| |
Collapse
|
2
|
Gruber RC, Wirak GS, Blazier AS, Lee L, Dufault MR, Hagan N, Chretien N, LaMorte M, Hammond TR, Cheong A, Ryan SK, Macklin A, Zhang M, Pande N, Havari E, Turner TJ, Chomyk A, Christie E, Trapp BD, Ofengeim D. BTK regulates microglial function and neuroinflammation in human stem cell models and mouse models of multiple sclerosis. Nat Commun 2024; 15:10116. [PMID: 39578444 PMCID: PMC11584639 DOI: 10.1038/s41467-024-54430-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
Neuroinflammation in the central nervous system (CNS), driven largely by resident phagocytes, has been proposed as a significant contributor to disability accumulation in multiple sclerosis (MS) but has not been addressed therapeutically. Bruton's tyrosine kinase (BTK) is expressed in both B-lymphocytes and innate immune cells, including microglia, where its role is poorly understood. BTK inhibition may provide therapeutic benefit within the CNS by targeting adaptive and innate immunity-mediated disease progression in MS. Using a CNS-penetrant BTK inhibitor (BTKi), we demonstrate robust in vivo effects in mouse models of MS. We further identify a BTK-dependent transcriptional signature in vitro, using the BTKi tolebrutinib, in mouse microglia, human induced pluripotent stem cell (hiPSC)-derived microglia, and a complex hiPSC-derived tri-culture system composed of neurons, astrocytes, and microglia, revealing modulation of neuroinflammatory pathways relevant to MS. Finally, we demonstrate that in MS tissue BTK is expressed in B-cells and microglia, with increased levels in lesions. Our data provide rationale for targeting BTK in the CNS to diminish neuroinflammation and disability accumulation.
Collapse
Affiliation(s)
- Ross C Gruber
- Sanofi, Cambridge, MA, USA
- Takeda, Cambridge, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | - Nilesh Pande
- Sanofi, Cambridge, MA, USA
- Voyager Therapeutics Inc, Cambridge, MA, USA
| | | | | | - Anthony Chomyk
- Department of Neurosciences, Cleveland Clinic, Cambridge, OH, USA
| | - Emilie Christie
- Department of Neurosciences, Cleveland Clinic, Cambridge, OH, USA
| | - Bruce D Trapp
- Department of Neurosciences, Cleveland Clinic, Cambridge, OH, USA
| | | |
Collapse
|
3
|
Cho Y, Cao Z, Luo X, Tian JJ, Hukkanen RR, Hussien R, Cancilla B, Chowdhury P, Li F, Ma S, LaGory EL, Schroeder M, Dusenberry A, Marshall L, Hawkins J, van Lookeren Campagne M, Zhou Y. NLRP10 maintains epidermal homeostasis by promoting keratinocyte survival and P63-dependent differentiation and barrier function. Cell Death Dis 2024; 15:759. [PMID: 39424623 PMCID: PMC11492288 DOI: 10.1038/s41419-024-07146-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/27/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Atopic dermatitis (AD) is a common chronic inflammatory skin disorder characterized by disrupted epidermal barrier function and aberrant immune responses. Despite recent developments in new therapeutics for AD, there is still a large unmet medical need for disease management due to the complex and multifactorial nature of AD. Recent genome-wide association studies (GWAS) have identified NLRP10 as a susceptible gene for AD but the physiological role of NLRP10 in skin homeostasis and AD remains unknown. Here we show that NLRP10 is downregulated in AD skin samples. Using an air-lift human skin equivalent culture, we demonstrate that NLRP10 promotes keratinocyte survival and is required for epidermal differentiation and barrier function. Mechanistically, NLRP10 limits cell death by preventing the recruitment of caspase-8 to the death inducing signaling complex (DISC) and by inhibiting its subsequent activation. NLRP10 also stabilizes p63, the master regulator of keratinocyte differentiation, to drive proper keratinocyte differentiation and to reinforce the barrier function. Our findings underscore NLRP10 as a key player in atopic dermatitis pathogenesis, highlighting NLRP10 as a potential target for therapeutic intervention to restore skin barrier function and homeostasis in AD.
Collapse
Affiliation(s)
- Yeonhee Cho
- Inflammation Research, Amgen Inc., South San Francisco, CA, USA
| | - Zhongzheng Cao
- Inflammation Research, Amgen Inc., South San Francisco, CA, USA
- Amgen R&D Postdoctoral Fellows Program, South San Francisco, CA, USA
| | - Xin Luo
- Center for Research Acceleration by Digital Innovation, Amgen Inc., South San Francisco, CA, USA
| | - Jennifer J Tian
- Translational Safety & Bioanalytical Sciences, Amgen Inc., South San Francisco, CA, USA
| | - Renee R Hukkanen
- Translational Safety & Bioanalytical Sciences, Amgen Inc, Cambridge, MA, USA
| | - Rajaa Hussien
- Translational Safety & Bioanalytical Sciences, Amgen Inc., South San Francisco, CA, USA
| | - Belinda Cancilla
- Translational Safety & Bioanalytical Sciences, Amgen Inc., South San Francisco, CA, USA
| | | | - Fei Li
- Structural Biology, Amgen Inc., South San Francisco, CA, USA
| | - Shining Ma
- Center for Research Acceleration by Digital Innovation, Amgen Inc., South San Francisco, CA, USA
| | - Edward L LaGory
- Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, CA, USA
| | - Mark Schroeder
- Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, CA, USA
| | | | | | - Jenn Hawkins
- Clinical Biomarkers, Amgen Inc, Thousand Oaks, CA, USA
| | | | - Yi Zhou
- Inflammation Research, Amgen Inc., South San Francisco, CA, USA.
| |
Collapse
|
4
|
Mantel N, Piras-Douce F, Chautard E, Marcos-Lopez E, Bodinham CL, Cosma A, Courtois V, Dhooge N, Gautheron S, Kaufmann SHE, Pizzoferro K, Lewis DJM, Martinon F, Pagnon A, Raynal F, Dereuddre-Bosquet N, Le Grand R. Cynomolgus macaques as a translational model of human immune responses to yellow fever 17D vaccination. J Virol 2024; 98:e0151623. [PMID: 38567951 PMCID: PMC11092345 DOI: 10.1128/jvi.01516-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/22/2023] [Indexed: 05/15/2024] Open
Abstract
The non-human primate (NHP) model (specifically rhesus and cynomolgus macaques) has facilitated our understanding of the pathogenic mechanisms of yellow fever (YF) disease and allowed the evaluation of the safety and efficacy of YF-17D vaccines. However, the accuracy of this model in mimicking vaccine-induced immunity in humans remains to be fully determined. We used a systems biology approach to compare hematological, biochemical, transcriptomic, and innate and antibody-mediated immune responses in cynomolgus macaques and human participants following YF-17D vaccination. Immune response progression in cynomolgus macaques followed a similar course as in adult humans but with a slightly earlier onset. Yellow fever virus neutralizing antibody responses occurred earlier in cynomolgus macaques [by Day 7[(D7)], but titers > 10 were reached in both species by D14 post-vaccination and were not significantly different by D28 [plaque reduction neutralization assay (PRNT)50 titers 3.6 Log vs 3.5 Log in cynomolgus macaques and human participants, respectively; P = 0.821]. Changes in neutrophils, NK cells, monocytes, and T- and B-cell frequencies were higher in cynomolgus macaques and persisted for 4 weeks versus less than 2 weeks in humans. Low levels of systemic inflammatory cytokines (IL-1RA, IL-8, MIP-1α, IP-10, MCP-1, or VEGF) were detected in either or both species but with no or only slight changes versus baseline. Similar changes in gene expression profiles were elicited in both species. These included enriched and up-regulated type I IFN-associated viral sensing, antiviral innate response, and dendritic cell activation pathways D3-D7 post-vaccination in both species. Hematological and blood biochemical parameters remained relatively unchanged versus baseline in both species. Low-level YF-17D viremia (RNAemia) was transiently detected in some cynomolgus macaques [28% (5/18)] but generally absent in humans [except one participant (5%; 1/20)].IMPORTANCECynomolgus macaques were confirmed as a valid surrogate model for replicating YF-17D vaccine-induced responses in humans and suggest a key role for type I IFN.
Collapse
Affiliation(s)
| | | | | | - Ernesto Marcos-Lopez
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay aux Roses, France
| | - Caroline L. Bodinham
- Surrey Clinical Research Centre, University of Surrey, Guildford, Surrey, United Kingdom
| | - Antonio Cosma
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay aux Roses, France
| | | | - Nina Dhooge
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay aux Roses, France
| | | | - Stefan H. E. Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany; Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, Texas, USA
| | - Kathleen Pizzoferro
- Surrey Clinical Research Centre, University of Surrey, Guildford, Surrey, United Kingdom
| | - David J. M. Lewis
- Surrey Clinical Research Centre, University of Surrey, Guildford, Surrey, United Kingdom
| | - Frédéric Martinon
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay aux Roses, France
| | - Anke Pagnon
- Research and Development, Sanofi, Marcy L'Etoile, France
| | - Franck Raynal
- Research and Development, Sanofi, Marcy L'Etoile, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay aux Roses, France
| | - Roger Le Grand
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay aux Roses, France
| |
Collapse
|
5
|
Anding A, Kinton S, Baranowski K, Brezzani A, De Busser H, Dufault MR, Finn P, Keefe K, Tetrault T, Li Y, Qiu W, Raes K, Vitse O, Zhang M, Ziegler R, Sardi SP, Hunter B, George K. Increasing Enzyme Mannose-6-Phosphate Levels but Not Miglustat Coadministration Enhances the Efficacy of Enzyme Replacement Therapy in Pompe Mice. J Pharmacol Exp Ther 2023; 387:188-203. [PMID: 37679046 DOI: 10.1124/jpet.123.001593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/17/2023] [Accepted: 06/01/2023] [Indexed: 09/09/2023] Open
Abstract
Pompe disease is a rare glycogen storage disorder caused by a deficiency in the lysosomal enzyme acid α-glucosidase, which leads to muscle weakness, cardiac and respiratory failure, and early mortality. Alglucosidase alfa, a recombinant human acid α-glucosidase, was the first approved treatment of Pompe disease, but its uptake into skeletal muscle via the cation-independent mannose-6-phosphate (M6P) receptor (CIMPR) is limited. Avalglucosidase alfa has received marketing authorization in several countries for infantile-onset and/or late-onset Pompe disease. This recently approved enzyme replacement therapy (ERT) was glycoengineered to maximize CIMPR binding through high-affinity interactions with ∼7 bis-M6P moieties. Recently, small molecules like the glucosylceramide synthase inhibitor miglustat were reported to increase the stability of recombinant human acid α-glucosidase, and it was suggested that an increased serum half-life would result in better glycogen clearance. Here, the effects of miglustat on alglucosidase alfa and avalglucosidase alfa stability, activity, and efficacy in Pompe mice were evaluated. Although miglustat increased the stability of both enzymes in fluorescent protein thermal shift assays and when incubated in neutral pH buffer over time, it reduced their enzymatic activity by ∼50%. Improvement in tissue glycogen clearance and transcriptional dysregulation in Pompe mice correlated with M6P levels but not with miglustat coadministration. These results further substantiate the crucial role of CIMPR binding in lysosomal targeting of ERTs. SIGNIFICANCE STATEMENT: This work describes important new insights into the treatment of Pompe disease using currently approved enzyme replacement therapies (ERTs) coadministered with miglustat. Although miglustat increased the stability of ERTs in vitro, there was no positive impact to glycogen clearance and transcriptional correction in Pompe mice. However, increasing mannose-6-phosphate levels resulted in increased cell uptake in vitro and increased glycogen clearance and transcriptional correction in Pompe mice, further underscoring the crucial role of cation-independent mannose-6-phosphate receptor-mediated lysosomal targeting for ERTs.
Collapse
Affiliation(s)
- Allyson Anding
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Sofia Kinton
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Kaitlyn Baranowski
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Alexander Brezzani
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Hilde De Busser
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Michael R Dufault
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Patrick Finn
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Kelly Keefe
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Tanya Tetrault
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Yi Li
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Weiliang Qiu
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Katrien Raes
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Olivier Vitse
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Mindy Zhang
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Robin Ziegler
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - S Pablo Sardi
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Bridge Hunter
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| | - Kelly George
- Metabolic and Lysosomal Storage Disease Research, Rare and Neurologic Diseases Therapeutic Area (A.A., S.K., K.B., A.B., P.F., K.K., T.T., R.Z., S.P.S., B.H., K.G.), Precision Medicine and Computational Biology (M.R.D., M.Z.), and Nonclinical Efficacy and Safety (W.Q.), Sanofi, Cambridge, Massachusetts; Manufacturing Sciences, Analytics, and Technology (MSAT), Sanofi, Geel, Belgium (H.D.B., K.R.); Medicinal Chemistry, Integrated Drug Discovery, Sanofi, Waltham, Massachusetts (Y.L.); and Pharmacokinetics Dynamics and Metabolism, Sanofi, Montpellier, France (O.V.)
| |
Collapse
|
6
|
Ma S, So M, Ghelani A, Srivas R, Sahoo A, Hall R, Liu W, Wu H, Yu S, Lu S, Song E, Cariaga T, Soto M, Zhou H, Li CM, Chaudhry A, Luo X, Sohn SJ. Attenuated IL-2 muteins leverage the TCR signal to enhance regulatory T cell homeostasis and response in vivo. Front Immunol 2023; 14:1257652. [PMID: 37809101 PMCID: PMC10556740 DOI: 10.3389/fimmu.2023.1257652] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/04/2023] [Indexed: 10/10/2023] Open
Abstract
Interleukin-2 (IL-2), along with T-cell receptor (TCR) signaling, are required to control regulatory T cell (Treg) homeostasis and function in vivo. Due to the heightened sensitivity to IL-2, Tregs retain the ability to respond to low-dose or attenuated forms of IL-2, as currently being developed for clinical use to treat inflammatory diseases. While attenuated IL-2 increases Treg selectivity, the question remains as to whether a weakened IL-2 signal sufficiently enhances Treg suppressive function(s) toward disease modification. To understand this question, we characterized the in vivo activity and transcriptomic profiles of two different attenuated IL-2 muteins in comparison with wildtype (WT) IL-2. Our study showed that, in addition to favoring Tregs, the attenuated muteins induced disproportionately robust effects on Treg activation and conversion to effector Treg (eTreg) phenotype. Our data furthermore suggested that Tregs activated by attenuated IL-2 muteins showed reduced dependence on TCR signal, at least in part due to the enhanced ability of IL-2 muteins to amplify the TCR signal in vivo. These results point to a new paradigm wherein IL-2 influences Tregs' sensitivity to antigenic signal, and that the combination effect may be leveraged for therapeutic use of attenuated IL-2 muteins.
Collapse
Affiliation(s)
- Shining Ma
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Michelle So
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Aazam Ghelani
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Rohith Srivas
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Anupama Sahoo
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Robyn Hall
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Wenjun Liu
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Hao Wu
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Sherman Yu
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Shiping Lu
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Elly Song
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Taryn Cariaga
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Marcus Soto
- Amgen Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Hong Zhou
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Chi-Ming Li
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | | | - Xin Luo
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Sue J. Sohn
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| |
Collapse
|
7
|
Righi M, Gannon I, Robson M, Srivastava S, Kokalaki E, Grothier T, Nannini F, Allen C, Bai YV, Sillibourne J, Cordoba S, Thomas S, Pule M. Enhancing CAR T-cell Therapy Using Fab-Based Constitutively Heterodimeric Cytokine Receptors. Cancer Immunol Res 2023; 11:1203-1221. [PMID: 37352396 PMCID: PMC10472109 DOI: 10.1158/2326-6066.cir-22-0640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 03/15/2023] [Accepted: 06/21/2023] [Indexed: 06/25/2023]
Abstract
Adoptive T-cell therapy aims to achieve lasting tumor clearance, requiring enhanced engraftment and survival of the immune cells. Cytokines are paramount modulators of T-cell survival and proliferation. Cytokine receptors signal via ligand-induced dimerization, and this principle has been hijacked utilizing nonnative dimerization domains. A major limitation of current technologies resides in the absence of a module that recapitulates the natural cytokine receptor heterodimeric pairing. To circumvent this, we created a new engineered cytokine receptor able to constitutively recreate receptor-heterodimer utilizing the heterodimerization domain derived from the IgG1 antibody (dFab_CCR). We found that the signal delivered by the dFab_CCR-IL2 proficiently mimicked the cytokine receptor heterodimerization, with transcriptomic signatures like those obtained by activation of the native IL2 receptor. Moreover, we found that this dimerization structure was agnostic, efficiently activating signaling through four cytokine receptor families. Using a combination of in vivo and in vitro screening approaches, we characterized a library of 18 dFab_CCRs coexpressed with a clinically relevant solid tumor-specific GD2-specific chimeric antigen receptor (CAR). Based on this characterization, we suggest that the coexpression of either the common β-chain GMCSF or the IL18 dFab_CCRs is optimal to improve CAR T-cell expansion, engraftment, and efficacy. Our results demonstrate how Fab dimerization is efficient and versatile in recapitulating a cytokine receptor heterodimerization signal. This module could be applied for the enhancement of adoptive T-cell therapies, as well as therapies based on other immune cell types. Furthermore, these results provide a choice of cytokine signal to incorporate with adoptive T-cell therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Francesco Nannini
- Department of Haematology, University College London, London, United Kingdom
| | | | | | | | | | | | - Martin Pule
- Autolus Therapeutics, London, United Kingdom
- Department of Haematology, University College London, London, United Kingdom
| |
Collapse
|
8
|
Zhang X, Hu LG, Lei Y, Stolina M, Homann O, Wang S, Véniant MM, Hsu YH. A transcriptomic and proteomic atlas of obesity and type 2 diabetes in cynomolgus monkeys. Cell Rep 2023; 42:112952. [PMID: 37556324 DOI: 10.1016/j.celrep.2023.112952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/16/2023] [Accepted: 07/23/2023] [Indexed: 08/11/2023] Open
Abstract
Obesity and type 2 diabetes (T2D) remain major global healthcare challenges, and developing therapeutics necessitates using nonhuman primate models. Here, we present a transcriptomic and proteomic atlas of all the major organs of cynomolgus monkeys with spontaneous obesity or T2D in comparison to healthy controls. Molecular changes occur predominantly in the adipose tissues of individuals with obesity, while extensive expression perturbations among T2D individuals are observed in many tissues such as the liver and kidney. Immune-response-related pathways are upregulated in obesity and T2D, whereas metabolism and mitochondrial pathways are downregulated. Moreover, we highlight some potential therapeutic targets, including SLC2A1 and PCSK1 in obesity as well as SLC30A8 and SLC2A2 in T2D. Our study provides a resource for exploring the complex molecular mechanism of obesity and T2D and developing therapies for these diseases, with limitations including lack of hypothalamus, isolated islets of Langerhans, longitudinal data, and body fat percentage.
Collapse
Affiliation(s)
- Xianglong Zhang
- Center for Research Acceleration by Digital Innovation (CRADI), Amgen Research, South San Francisco, CA 94080, USA
| | | | - Ying Lei
- Research China, Amgen Research, Shanghai 200020, China
| | - Marina Stolina
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA 91320, USA
| | - Oliver Homann
- Center for Research Acceleration by Digital Innovation (CRADI), Amgen Research, South San Francisco, CA 94080, USA
| | - Songli Wang
- Research Biomics, Amgen Research, South San Francisco, CA 94080, USA
| | - Murielle M Véniant
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA 91320, USA.
| | - Yi-Hsiang Hsu
- Marcus Institute for Aging Research and Harvard Medical School, Boston, MA 02131, USA.
| |
Collapse
|
9
|
Han Z, Wu X, Qin H, Yuan YC, Schmolze D, Su C, Zain J, Moyal L, Hodak E, Sanchez JF, Lee PP, Feng M, Rosen ST, Querfeld C. Reprogramming of PD-1+ M2-like tumor-associated macrophages with anti-PD-L1 and lenalidomide in cutaneous T cell lymphoma. JCI Insight 2023; 8:e163518. [PMID: 37427589 PMCID: PMC10371344 DOI: 10.1172/jci.insight.163518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 05/19/2023] [Indexed: 07/11/2023] Open
Abstract
Cutaneous T cell lymphoma (CTCL) is a disfiguring and incurable disease characterized by skin-homing malignant T cells surrounded by immune cells that promote CTCL growth through an immunosuppressive tumor microenvironment (TME). Preliminary data from our phase I clinical trial of anti-programmed cell death ligand 1 (anti-PD-L1) combined with lenalidomide in patients with relapsed/refractory CTCL demonstrated promising clinical efficacy. In the current study, we analyzed the CTCL TME, which revealed a predominant PD-1+ M2-like tumor-associated macrophage (TAM) subtype with upregulated NF-κB and JAK/STAT signaling pathways and an aberrant cytokine and chemokine profile. Our in vitro studies investigated the effects of anti-PD-L1 and lenalidomide on PD-1+ M2-like TAMs. The combinatorial treatment synergistically induced functional transformation of PD-1+ M2-like TAMs toward a proinflammatory M1-like phenotype that gained phagocytic activity upon NF-κB and JAK/STAT inhibition, altered their migration through chemokine receptor alterations, and stimulated effector T cell proliferation. Lenalidomide was more effective than anti-PD-L1 in downregulation of the immunosuppressive IL-10, leading to decreased expression of both PD-1 and PD-L1. Overall, PD-1+ M2-like TAMs play an immunosuppressive role in CTCL. Anti-PD-L1 combined with lenalidomide provides a therapeutic strategy to enhance antitumor immunity by targeting PD-1+ M2-like TAMs in the CTCL TME.
Collapse
Affiliation(s)
- Zhen Han
- Division of Dermatology
- Beckman Research Institute
| | - Xiwei Wu
- Department of Computational and Quantitative Medicine
- Integrative Genomics Core
| | - Hanjun Qin
- Department of Computational and Quantitative Medicine
| | - Yate-Ching Yuan
- Department of Computational and Quantitative Medicine
- Center for informatics
| | | | - Chingyu Su
- Division of Dermatology
- Beckman Research Institute
| | - Jasmine Zain
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Lilach Moyal
- Department of Dermatology, Rabin Medical Center, Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Emmilia Hodak
- Department of Dermatology, Rabin Medical Center, Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv, Israel
- Beilinson Hospital, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - James F Sanchez
- Beckman Research Institute
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Peter P Lee
- Beckman Research Institute
- Department of Immuno-Oncology, City of Hope, Duarte, California, USA
| | - Mingye Feng
- Beckman Research Institute
- Department of Immuno-Oncology, City of Hope, Duarte, California, USA
| | - Steven T Rosen
- Beckman Research Institute
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | - Christiane Querfeld
- Division of Dermatology
- Beckman Research Institute
- Department of Pathology, and
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| |
Collapse
|
10
|
Shitara K, Di Bartolomeo M, Mandala M, Ryu MH, Caglevic C, Olesinski T, Chung HC, Muro K, Goekkurt E, McDermott RS, Mansoor W, Wainberg ZA, Shih CS, Kobie J, Nebozhyn M, Cristescu R, Cao ZA, Loboda A, Özgüroğlu M. Association between gene expression signatures and clinical outcomes of pembrolizumab versus paclitaxel in advanced gastric cancer: exploratory analysis from the randomized, controlled, phase III KEYNOTE-061 trial. J Immunother Cancer 2023; 11:e006920. [PMID: 37399357 PMCID: PMC10314681 DOI: 10.1136/jitc-2023-006920] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND In the randomized, controlled, phase III KEYNOTE-061 trial, second-line pembrolizumab did not significantly prolong overall survival (OS) versus paclitaxel in patients with PD-L1-positive (combined positive score ≥1) advanced gastric/gastroesophageal junction (G/GEJ) cancer but did elicit a longer duration of response and offered a favorable safety profile. This prespecified exploratory analysis was conducted to evaluate associations between tumor gene expression signatures and clinical outcomes in the phase III KEYNOTE-061 trial. METHODS Using RNA sequencing data obtained from formalin-fixed, paraffin-embedded baseline tumor tissue samples, we evaluated the 18-gene T-cell-inflamed gene expression profile (TcellinfGEP) and 10 non-TcellinfGEP signatures (angiogenesis, glycolysis, granulocytic myeloid-derived suppressor cell (gMDSC), hypoxia, monocytic MDSC (mMDSC), MYC, proliferation, RAS, stroma/epithelial-to-mesenchymal transition/transforming growth factor-β, WNT). The association between each signature on a continuous scale and outcomes was analyzed using logistic (objective response rate (ORR)) and Cox proportional hazards regression (progression-free survival (PFS) and OS). One-sided (pembrolizumab) and two-sided (paclitaxel) p values were calculated for TcellinfGEP (prespecified α=0.05) and the 10 non-TcellinfGEP signatures (multiplicity-adjusted; prespecified α=0.10). RESULTS RNA sequencing data were available for 137 patients in each treatment group. TcellinfGEP was positively associated with ORR (p=0.041) and PFS (p=0.026) for pembrolizumab but not paclitaxel (p>0.05). The TcellinfGEP-adjusted mMDSC signature was negatively associated with ORR (p=0.077), PFS (p=0.057), and OS (p=0.033) for pembrolizumab, while the TcellinfGEP-adjusted glycolysis (p=0.018), MYC (p=0.057), and proliferation (p=0.002) signatures were negatively associated with OS for paclitaxel. CONCLUSIONS This exploratory analysis of tumor TcellinfGEP showed associations with ORR and PFS for pembrolizumab but not for paclitaxel. TcellinfGEP-adjusted mMDSC signature was negatively associated with ORR, PFS, and OS for pembrolizumab but not paclitaxel. These data suggest myeloid-driven suppression may play a role in resistance to PD-1 inhibition in G/GEJ cancer and support a strategy of considering immunotherapy combinations which target this myeloid axis. TRIAL REGISTRATION NUMBER NCT02370498.
Collapse
Affiliation(s)
- Kohei Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Mario Mandala
- Unit of Medical Oncology, University of Perugia, Perugia, Italy
| | - Min-Hee Ryu
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea (the Republic of)
| | - Christian Caglevic
- Cancer Research Department, Instituto Oncologico Fundacion Arturo Lopez Perez, Santiago, Chile
| | - Tomasz Olesinski
- Department of Gastrointestinal Cancers and Neuroendocrine Tumors Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Hyun Cheol Chung
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Eray Goekkurt
- Hematology-Oncology Practice Eppendorf (HOPE) and University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Wasat Mansoor
- Christie Hospital NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Zev A Wainberg
- Division of Hematology/Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | | | | | | | | | | | | | - Mustafa Özgüroğlu
- Division of Medical Oncology, Clinical Trial Unit, Istanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
11
|
Luo W, Adamska JZ, Li C, Verma R, Liu Q, Hagan T, Wimmers F, Gupta S, Feng Y, Jiang W, Zhou J, Valore E, Wang Y, Trisal M, Subramaniam S, Osborne TF, Pulendran B. SREBP signaling is essential for effective B cell responses. Nat Immunol 2023; 24:337-348. [PMID: 36577930 PMCID: PMC10928801 DOI: 10.1038/s41590-022-01376-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 10/31/2022] [Indexed: 12/29/2022]
Abstract
Our previous study using systems vaccinology identified an association between the sterol regulatory binding protein (SREBP) pathway and humoral immune response to vaccination in humans. To investigate the role of SREBP signaling in modulating immune responses, we generated mice with B cell- or CD11c+ antigen-presenting cell (APC)-specific deletion of SCAP, an essential regulator of SREBP signaling. Ablation of SCAP in CD11c+ APCs had no effect on immune responses. In contrast, SREBP signaling in B cells was critical for antibody responses, as well as the generation of germinal centers,memory B cells and bone marrow plasma cells. SREBP signaling was required for metabolic reprogramming in activated B cells. Upon mitogen stimulation, SCAP-deficient B cells could not proliferate and had decreased lipid rafts. Deletion of SCAP in germinal center B cells using AID-Cre decreased lipid raft content and cell cycle progression. These studies provide mechanistic insights coupling sterol metabolism with the quality and longevity of humoral immunity.
Collapse
Affiliation(s)
- Wei Luo
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Julia Z Adamska
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Chunfeng Li
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Rohit Verma
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Qing Liu
- Department of Biological Sciences, Clemson University, Clemson, SC, USA
| | - Thomas Hagan
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Florian Wimmers
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Molecular Medicine, Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany
| | - Shakti Gupta
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
| | - Yupeng Feng
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Wenxia Jiang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jiehao Zhou
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Erika Valore
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Yanli Wang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Meera Trisal
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Shankar Subramaniam
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
| | - Timothy F Osborne
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
12
|
Gupta S, Polit LD, Fitzgerald M, Rowland HA, Murali D, Buckley NJ, Subramaniam S. Temporal transcriptional control of neural induction in human induced pluripotent stem cells. Front Mol Neurosci 2023; 16:1139287. [PMID: 37213689 PMCID: PMC10195998 DOI: 10.3389/fnmol.2023.1139287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/14/2023] [Indexed: 05/23/2023] Open
Abstract
Introduction Neural induction of human induced pluripotent stem cells represents a critical switch in cell state during which pluripotency is lost and commitment to a neural lineage is initiated. Although many of the key transcription factors involved in neural induction are known, we know little of the temporal and causal relationships that are required for this state transition. Methods Here, we have carried out a longitudinal analysis of the transcriptome of human iPSCs undergoing neural induction. Using the temporal relationships between the changing profile of key transcription factors and subsequent changes in their target gene expression profiles, we have identified distinct functional modules operative throughout neural induction. Results In addition to modules that govern loss of pluripotency and gain of neural ectoderm identity, we discover other modules governing cell cycle and metabolism. Strikingly, some of these functional modules are retained throughout neural induction, even though the gene membership of the module changes. Systems analysis identifies other modules associated with cell fate commitment, genome integrity, stress response and lineage specification. We then focussed on OTX2, one of the most precociously activated transcription factors during neural induction. Our temporal analysis of OTX2 target gene expression identified several OTX2 regulated gene modules representing protein remodelling, RNA splicing and RNA processing. Further CRISPRi inhibition of OTX2 prior to neural induction promotes an accelerated loss of pluripotency and a precocious and aberrant neural induction disrupting some of the previously identified modules. Discussion We infer that OTX2 has a diverse role during neural induction and regulates many of the biological processes that are required for loss of pluripotency and gain of neural identity. This dynamical analysis of transcriptional changes provides a unique perspective of the widespread remodelling of the cell machinery that occurs during neural induction of human iPSCs.
Collapse
Affiliation(s)
- Shakti Gupta
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Lucia Dutan Polit
- Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
| | - Michael Fitzgerald
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Helen A. Rowland
- Department of Psychiatry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, United Kingdom
| | - Divya Murali
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Noel J. Buckley
- Department of Psychiatry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, United Kingdom
- *Correspondence: Noel J. Buckley, ; Shankar Subramaniam,
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
- Departments of Computer Science and Engineering, and Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, United States
- *Correspondence: Noel J. Buckley, ; Shankar Subramaniam,
| |
Collapse
|
13
|
Multi-omics profiling of collagen-induced arthritis mouse model reveals early metabolic dysregulation via SIRT1 axis. Sci Rep 2022; 12:11830. [PMID: 35821263 PMCID: PMC9276706 DOI: 10.1038/s41598-022-16005-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/04/2022] [Indexed: 11/24/2022] Open
Abstract
Rheumatoid arthritis (RA) is characterized by joint infiltration of immune cells and synovial inflammation which leads to progressive disability. Current treatments improve the disease outcome, but the unmet medical need is still high. New discoveries over the last decade have revealed the major impact of cellular metabolism on immune cell functions. So far, a comprehensive understanding of metabolic changes during disease development, especially in the diseased microenvironment, is still limited. Therefore, we studied the longitudinal metabolic changes during the development of murine arthritis by integrating metabolomics and transcriptomics data. We identified an early change in macrophage pathways which was accompanied by oxidative stress, a drop in NAD+ level and induction of glucose transporters. We discovered inhibition of SIRT1, a NAD-dependent histone deacetylase and confirmed its dysregulation in human macrophages and synovial tissues of RA patients. Mining this database should enable the discovery of novel metabolic targets and therapy opportunities in RA.
Collapse
|
14
|
Shui IM, Liu XQ, Zhao Q, Kim ST, Sun Y, Yearley JH, Choudhury T, Webber AL, Krepler C, Cristescu R, Lee J. Baseline and post-treatment biomarkers of resistance to anti-PD-1 therapy in acral and mucosal melanoma: an observational study. J Immunother Cancer 2022; 10:jitc-2022-004879. [PMID: 35793874 PMCID: PMC9260847 DOI: 10.1136/jitc-2022-004879] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2022] [Indexed: 12/14/2022] Open
Abstract
Background Immunotherapies targeting programmed cell death-1 (PD-1) and its ligands have improved clinical outcomes for advanced melanoma. However, many tumors exhibit primary resistance or acquire secondary resistance after an initial positive response. The mechanisms of resistance are not well understood, and no validated predictive biomarkers are available. This exploratory study aimed to characterize baseline differences and molecular changes arising during treatment in acral and mucosal melanomas that exhibited primary or secondary resistance to anti-PD-1 monotherapy. Methods This was an observational retrospective study of 124 patients who had been treated for metastatic acral or mucosal melanoma with anti-PD-1 monotherapy. Tumor samples were collected at baseline (all patients) and post-treatment (resistant tumors only) and were assayed by immunohistochemistry, whole-exome sequencing, and RNA sequencing. Results At baseline, more non-progressor than resistant tumors exhibited expression of PD-L1, immune cell infiltration, and high tumor mutational burden (TMB); baseline PD-L1 expression was also more common in secondary-resistant than in primary-resistant tumors as well as in late versus early secondary-resistant tumors. Non-progressor tumors also had higher median baseline expression of an 18-gene T cell-inflamed gene expression profile (TcellinfGEP). Among resistant tumors, the proportion of PD-L1-positive melanomas and the expression of the TcellinfGEP mRNA signature increased during treatment, while the expression of mRNA signatures related to WNT and INFA1 signaling decreased. There was evidence for greater changes from baseline in secondary-resistant versus primary-resistant tumors for some markers, including expression of RAS-related and WNT-related mRNA signatures and density of CD11c+ and FOXP3+ T cells. Greater changes in CD11c+ cell density were observed in early compared with late secondary-resistant tumors. Conclusions Our findings suggest that TcellinfGEP and PD-L1 expression, TMB, immune cell infiltration, and RAS and WNT signaling warrant further investigation as potential mechanisms and/or biomarkers of anti-PD-1 therapy resistance in acral and mucosal melanomas. Confirmation of these findings in larger populations is needed.
Collapse
Affiliation(s)
| | | | - Qing Zhao
- Merck & Co., Inc, Rahway, New Jersey, USA
| | - Seung Tae Kim
- Hematology and Oncology, Samsung Medical Center, Gangnam-gu, South Korea
| | - Yuan Sun
- Merck & Co., Inc, Rahway, New Jersey, USA
| | | | | | | | | | | | - Jeeyun Lee
- Hematology and Oncology, Samsung Medical Center, Gangnam-gu, South Korea
| |
Collapse
|
15
|
Cristescu R, Nebozhyn M, Zhang C, Albright A, Kobie J, Huang L, Zhao Q, Wang A, Ma H, Alexander Cao Z, Morrissey M, Ribas A, Grivas P, Cescon DW, McClanahan TK, Snyder A, Ayers M, Lunceford J, Loboda A. Transcriptomic Determinants of Response to Pembrolizumab Monotherapy across Solid Tumor Types. Clin Cancer Res 2022; 28:1680-1689. [PMID: 34965943 PMCID: PMC9762333 DOI: 10.1158/1078-0432.ccr-21-3329] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/12/2021] [Accepted: 12/20/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE To explore relationships between biological gene expression signatures and pembrolizumab response. EXPERIMENTAL DESIGN RNA-sequencing data on baseline tumor tissue from 1,188 patients across seven tumor types treated with pembrolizumab monotherapy in nine clinical trials were used. A total of 11 prespecified gene expression signatures [18-gene T-cell-inflamed gene expression profile (TcellinfGEP), angiogenesis, hypoxia, glycolysis, proliferation, MYC, RAS, granulocytic myeloid-derived suppressor cell (gMDSC), monocytic myeloid-derived suppressor cell (mMDSC), stroma/epithelial-to-mesenchymal transition (EMT)/TGFβ, and WNT] were evaluated for their relationship to objective response rate (per RECIST, version 1.1). Logistic regression analysis of response for consensus signatures was adjusted for tumor type, Eastern Cooperative Oncology Group performance status, and TcellinfGEP, an approach equivalent to evaluating the association between response and the residuals of consensus signatures after detrending them for their relationship with the TcellinfGEP (previously identified as a determinant of pembrolizumab response) and tumor type. Testing of the 10 prespecified non-TcellinfGEP consensus signatures for negative association [except proliferation (hypothesized positive association)] with response was adjusted for multiplicity. RESULTS Covariance patterns of the 11 signatures (including TcellinfGEP) identified in Merck-Moffitt and The Cancer Genome Atlas datasets showed highly concordant coexpression patterns in the RNA-sequencing data from pembrolizumab trials. TcellinfGEP was positively associated with response; signatures for angiogenesis, mMDSC, and stroma/EMT/TGFβ were negatively associated with response to pembrolizumab monotherapy. CONCLUSIONS These findings suggest that features beyond IFNγ-related T-cell inflammation may be relevant to anti-programmed death 1 monotherapy response and may define other axes of tumor biology as candidates for pembrolizumab combinations. See related commentary by Cho et al., p. 1479.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qing Zhao
- Merck & Co., Inc., Kenilworth, New Jersey
| | - Anran Wang
- Merck & Co., Inc., Kenilworth, New Jersey
| | - Hua Ma
- Merck & Co., Inc., Kenilworth, New Jersey
| | | | | | - Antoni Ribas
- Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Petros Grivas
- University of Washington Seattle Cancer Care Alliance and Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - David W. Cescon
- UHN Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | | | - Mark Ayers
- Merck & Co., Inc., Kenilworth, New Jersey
| | | | - Andrey Loboda
- Merck & Co., Inc., Kenilworth, New Jersey.,Corresponding Author: Andrey Loboda, Genetics and Pharmacogenomics, Merck & Co., Inc., Boston, MA 02115. Phone: 617-835-7783; E-mail:
| |
Collapse
|
16
|
Differential interferon-α subtype induced immune signatures are associated with suppression of SARS-CoV-2 infection. Proc Natl Acad Sci U S A 2022; 119:2111600119. [PMID: 35131898 PMCID: PMC8872780 DOI: 10.1073/pnas.2111600119] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Type I interferons (IFN-I) exhibit various biological effects during viral infections, and they have been successfully used for clinical treatment of viral diseases. Humans express 12 IFNα subtypes, which strongly differ in their antiviral responses against different viruses. Here we analyzed the antiviral activity of all human IFNα subtypes against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to identify the underlying immune signatures and explore their therapeutic potential. Our data provide a systemic pattern of antiviral host effector responses mediated by high antiviral IFN-I, which could help to identify key cellular effectors targeted in novel therapeutic approaches against SARS-CoV-2 infection. Type I interferons (IFN-I) exert pleiotropic biological effects during viral infections, balancing virus control versus immune-mediated pathologies, and have been successfully employed for the treatment of viral diseases. Humans express 12 IFN-alpha (α) subtypes, which activate downstream signaling cascades and result in distinct patterns of immune responses and differential antiviral responses. Inborn errors in IFN-I immunity and the presence of anti-IFN autoantibodies account for very severe courses of COVID-19; therefore, early administration of IFN-I may be protective against life-threatening disease. Here we comprehensively analyzed the antiviral activity of all IFNα subtypes against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to identify the underlying immune signatures and explore their therapeutic potential. Prophylaxis of primary human airway epithelial cells (hAEC) with different IFNα subtypes during SARS-CoV-2 infection uncovered distinct functional classes with high, intermediate, and low antiviral IFNs. In particular, IFNα5 showed superior antiviral activity against SARS-CoV-2 infection in vitro and in SARS-CoV-2–infected mice in vivo. Dose dependency studies further displayed additive effects upon coadministration with the broad antiviral drug remdesivir in cell culture. Transcriptomic analysis of IFN-treated hAEC revealed different transcriptional signatures, uncovering distinct, intersecting, and prototypical genes of individual IFNα subtypes. Global proteomic analyses systematically assessed the abundance of specific antiviral key effector molecules which are involved in IFN-I signaling pathways, negative regulation of viral processes, and immune effector processes for the potent antiviral IFNα5. Taken together, our data provide a systemic, multimodular definition of antiviral host responses mediated by defined IFN-I. This knowledge will support the development of novel therapeutic approaches against SARS-CoV-2.
Collapse
|
17
|
Rubel D, Boulanger J, Craciun F, Xu EY, Zhang Y, Phillips L, Callahan M, Weber W, Song W, Ngai N, Bukanov NO, Shi X, Hariri A, Husson H, Ibraghimov-Beskrovnaya O, Liu S, Gross O. Anti-microRNA-21 Therapy on Top of ACE Inhibition Delays Renal Failure in Alport Syndrome Mouse Models. Cells 2022; 11:cells11040594. [PMID: 35203245 PMCID: PMC8869926 DOI: 10.3390/cells11040594] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 02/01/2023] Open
Abstract
Col4a3−/− Alport mice serve as an animal model for renal fibrosis. MicroRNA-21 (miR-21) expression has been shown to be increased in the kidneys of Alport syndrome patients. Here, we investigated the nephroprotective effects of Lademirsen anti-miR-21 therapy. We used a fast-progressing Col4a3−/− mouse model with a 129/SvJ background and an intermediate-progressing F1 hybrid mouse model with a mixed genetic background, with angiotensin-converting enzyme inhibitor (ACEi) monotherapy in combination with anti-miR-21 therapy. In the fast-progressing model, the anti miR-21 and ACEi therapies showed an additive effect in the reduction in fibrosis, the decline of proteinuria, the preservation of kidney function and increased survival. In the intermediate-progressing F1 model, the anti-miR-21 and ACEi therapies individually improved kidney pathology. Both also improved kidney function and survival; however, the combination showed a significant additive effect, particularly for survival. RNA sequencing (RNA-seq) gene expression profiling revealed that the anti-miR-21 and ACEi therapies modulate several common pathways. However, anti-miR-21 was particularly effective at normalizing the expression profiles of the genes involved in renal tubulointerstitial injury pathways. In conclusion, significant additive effects were detected for the combination of anti-miR-21 and ACEi therapies on kidney function, pathology and survival in Alport mouse models, as well as a strong differential effect of anti-miR-21 on the renal expression of fibrotic factors. These results support the addition of anti-miR-21 to the current standard of care (ACEi) in ongoing clinical trials in patients with Alport syndrome.
Collapse
Affiliation(s)
- Diana Rubel
- Clinic for Nephrology and Rheumatology, University Medical Center Goettingen, 37075 Goettingen, Germany; (D.R.); (Y.Z.)
| | | | - Florin Craciun
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
| | - Ethan Y. Xu
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
- Excision BioTherapeutics, San Francisco, CA 94111, USA
| | - Yanqin Zhang
- Clinic for Nephrology and Rheumatology, University Medical Center Goettingen, 37075 Goettingen, Germany; (D.R.); (Y.Z.)
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Lucy Phillips
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
- Abbvie Bioresearch Center, Worcester, MA 01605, USA
| | - Michelle Callahan
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
| | - William Weber
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
- Takeda Pharmaceuticals, Cambridge, MA 02139, USA
| | - Wenping Song
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
| | - Nicholas Ngai
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
| | - Nikolay O. Bukanov
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
- Janssen Pharmaceuticals, Boston, MA 02115, USA
| | - Xingyi Shi
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
- Novartis Institute for BioMedical Research, Boston, MA 02139, USA
| | - Ali Hariri
- Sanofi-Genzyme, Clinical Development, Cambridge, MA 02142, USA; (A.H.); (S.L.)
- Eloxx Pharmaceuticals, Watertown, MA 02140, USA
| | - Hervé Husson
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
| | - Oxana Ibraghimov-Beskrovnaya
- Sanofi-Genzyme Research and Development, Framingham, MA 02118, USA; (F.C.); (E.Y.X.); (L.P.); (M.C.); (W.W.); (W.S.); (N.N.); (N.O.B.); (X.S.); (H.H.); (O.I.-B.)
- Dyne Therapeutics, Waltham, MA 02451, USA
| | - Shiguang Liu
- Sanofi-Genzyme, Clinical Development, Cambridge, MA 02142, USA; (A.H.); (S.L.)
| | - Oliver Gross
- Clinic for Nephrology and Rheumatology, University Medical Center Goettingen, 37075 Goettingen, Germany; (D.R.); (Y.Z.)
- Correspondence: ; Tel.: +49-551-39-60488
| |
Collapse
|
18
|
Paavola KJ, Roda JM, Lin VY, Chen P, O'Hollaren KP, Ventura R, Crawley SC, Li B, Chen HIH, Malmersjö S, Sharkov NA, Horner G, Guo W, Kutach AK, Mondal K, Zhang Z, Lichtman JS, Song C, Rivera LB, Liu W, Luo J, Wang Y, Solloway MJ, Allan BB, Kekatpure A, Starck SR, Haldankar R, Fan B, Chu C, Tang J, Molgora M, Colonna M, Kaplan DD, Hsu JY. The Fibronectin-ILT3 Interaction Functions as a Stromal Checkpoint that Suppresses Myeloid Cells. Cancer Immunol Res 2021; 9:1283-1297. [PMID: 34426457 PMCID: PMC9414285 DOI: 10.1158/2326-6066.cir-21-0240] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/09/2021] [Accepted: 08/17/2021] [Indexed: 01/07/2023]
Abstract
Suppressive myeloid cells inhibit antitumor immunity by preventing T-cell responses. Immunoglobulin-like transcript 3 (ILT3; also known as LILRB4) is highly expressed on tumor-associated myeloid cells and promotes their suppressive phenotype. However, the ligand that engages ILT3 within the tumor microenvironment and renders tumor-associated myeloid cells suppressive is unknown. Using a screening approach, we identified fibronectin as a functional ligand for ILT3. The interaction of fibronectin with ILT3 polarized myeloid cells toward a suppressive state, and these effects were reversed with an ILT3-specific antibody that blocked the interaction of ILT3 with fibronectin. Furthermore, ex vivo treatment of human tumor explants with anti-ILT3 reprogrammed tumor-associated myeloid cells toward a stimulatory phenotype. Thus, the ILT3-fibronectin interaction represents a "stromal checkpoint" through which the extracellular matrix actively suppresses myeloid cells. By blocking this interaction, tumor-associated myeloid cells may acquire a stimulatory phenotype, potentially resulting in increased antitumor T-cell responses.
Collapse
Affiliation(s)
| | - Julie M. Roda
- NGM Biopharmaceuticals, South San Francisco, California
| | - Vicky Y. Lin
- NGM Biopharmaceuticals, South San Francisco, California
| | - Peirong Chen
- NGM Biopharmaceuticals, South San Francisco, California
| | | | | | | | - Betty Li
- NGM Biopharmaceuticals, South San Francisco, California
| | | | | | | | | | - Wei Guo
- NGM Biopharmaceuticals, South San Francisco, California
| | | | | | - Zhen Zhang
- NGM Biopharmaceuticals, South San Francisco, California
| | | | | | - Lee B. Rivera
- NGM Biopharmaceuticals, South San Francisco, California
| | - Wenhui Liu
- NGM Biopharmaceuticals, South San Francisco, California
| | - Jian Luo
- NGM Biopharmaceuticals, South San Francisco, California
| | - Yan Wang
- NGM Biopharmaceuticals, South San Francisco, California
| | | | | | | | | | - Raj Haldankar
- NGM Biopharmaceuticals, South San Francisco, California
| | - Bin Fan
- NGM Biopharmaceuticals, South San Francisco, California
| | - Chun Chu
- NGM Biopharmaceuticals, South San Francisco, California
| | - Jie Tang
- NGM Biopharmaceuticals, South San Francisco, California
| | - Martina Molgora
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Jer-Yuan Hsu
- NGM Biopharmaceuticals, South San Francisco, California.,Corresponding Author: Jer-Yuan Hsu, NGM Biopharmaceuticals, 333 Oyster Point Boulevard, South San Francisco, CA 94080. Phone: 650-243-5579; Fax: 650-583-1646; E-mail:
| |
Collapse
|
19
|
Raghunandan S, Ramachandran S, Ke E, Miao Y, Lal R, Chen ZB, Subramaniam S. Heme Oxygenase-1 at the Nexus of Endothelial Cell Fate Decision Under Oxidative Stress. Front Cell Dev Biol 2021; 9:702974. [PMID: 34595164 PMCID: PMC8476872 DOI: 10.3389/fcell.2021.702974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/17/2021] [Indexed: 12/31/2022] Open
Abstract
Endothelial cells (ECs) form the inner lining of blood vessels and are central to sensing chemical perturbations that can lead to oxidative stress. The degree of stress is correlated with divergent phenotypes such as quiescence, cell death, or senescence. Each possible cell fate is relevant for a different aspect of endothelial function, and hence, the regulation of cell fate decisions is critically important in maintaining vascular health. This study examined the oxidative stress response (OSR) in human ECs at the boundary of cell survival and death through longitudinal measurements, including cellular, gene expression, and perturbation measurements. 0.5 mM hydrogen peroxide (HP) produced significant oxidative stress, placed the cell at this junction, and provided a model to study the effectors of cell fate. The use of systematic perturbations and high-throughput measurements provide insights into multiple regimes of the stress response. Using a systems approach, we decipher molecular mechanisms across these regimes. Significantly, our study shows that heme oxygenase-1 (HMOX1) acts as a gatekeeper of cell fate decisions. Specifically, HP treatment of HMOX1 knockdown cells reversed the gene expression of about 51% of 2,892 differentially expressed genes when treated with HP alone, affecting a variety of cellular processes, including anti-oxidant response, inflammation, DNA injury and repair, cell cycle and growth, mitochondrial stress, metabolic stress, and autophagy. Further analysis revealed that these switched genes were highly enriched in three spatial locations viz., cell surface, mitochondria, and nucleus. In particular, it revealed the novel roles of HMOX1 on cell surface receptors EGFR and IGFR, mitochondrial ETCs (MTND3, MTATP6), and epigenetic regulation through chromatin modifiers (KDM6A, RBBP5, and PPM1D) and long non-coding RNA (lncRNAs) in orchestrating the cell fate at the boundary of cell survival and death. These novel aspects suggest that HMOX1 can influence transcriptional and epigenetic modulations to orchestrate OSR affecting cell fate decisions.
Collapse
Affiliation(s)
- Sindhushree Raghunandan
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Srinivasan Ramachandran
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Eugene Ke
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Yifei Miao
- Department of Diabetes Complications and Metabolism, City of Hope, Duarte, CA, United States
| | - Ratnesh Lal
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States.,Department of Mechanical and Aerospace Engineering, University of California, San Diego, San Diego, CA, United States
| | - Zhen Bouman Chen
- Department of Diabetes Complications and Metabolism, City of Hope, Duarte, CA, United States
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States.,Department of Computer Science and Engineering, University of California, San Diego, San Diego, CA, United States.,Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
20
|
Buchheim JI, Billaud JN, Feuerecker M, Strewe C, Dangoisse C, Osterman A, Mehta S, Crucian B, Schelling G, Choukér A. Exploratory RNA-seq analysis in healthy subjects reveals vulnerability to viral infections during a 12- month period of isolation and confinement. Brain Behav Immun Health 2021; 9:100145. [PMID: 34589891 PMCID: PMC8474453 DOI: 10.1016/j.bbih.2020.100145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/07/2020] [Accepted: 09/16/2020] [Indexed: 11/28/2022] Open
Abstract
Exposure to stressful environments weakens immunity evidenced by a detectable reactivation of dormant viruses. The mechanism behind this observation remains unclear. We performed next generation sequencing from RNA extracted from blood samples of 8 male subjects collected before, during and after a 12-month stay at the Antarctic station Concordia. RNA-seq data analysis was done using QIAGEN Ingenuity Pathway Analysis (IPA) software. Data revealed the inactivation of key immune functions such as chemotaxis and leukocyte recruitment which persisted after return. Next to the activation of the stress response eIF2 pathway, interferon signaling was predicted inactivated due to a downregulation of 14 downstream genes involved in antiviral immunity. Among them, the interferon stimulated genes (ISGs) IFITM2 and 3 as well as IFIT3 exhibited the strongest fold changes and IFIT3 remained downregulated even after return. Impairment of antiviral immunity in winter-over crew can be explained by the downregulation of a battery of ISGs. Whole blood transcriptome analysis during 12-months of isolation in the Antarctic. Data show an inactivation of key immune functions and pathways without recovery. The IFN pathway is most affected showing a downregulation of 14 downstream genes. The results suggest impairment of antiviral immunity and vulnerability to infection.
Collapse
Affiliation(s)
- Judith-Irina Buchheim
- Laboratory of Translational Research "Stress and Immunity", Department of Anesthesiology, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Marchioninistr. 15, 81377, Munich, Germany
| | | | - Matthias Feuerecker
- Laboratory of Translational Research "Stress and Immunity", Department of Anesthesiology, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Marchioninistr. 15, 81377, Munich, Germany
| | - Claudia Strewe
- Laboratory of Translational Research "Stress and Immunity", Department of Anesthesiology, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Marchioninistr. 15, 81377, Munich, Germany
| | - Carole Dangoisse
- Department of Anesthesia and Critical Care, Ysbyty Gwynedd Hospital, Bangor, Wales, UK
| | - Andreas Osterman
- Max von Pettenkofer Institute, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Germany
| | | | | | - Gustav Schelling
- Department of Anesthesiology, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Marchioninistr. 15, 81377, Munich, Germany
| | - Alexander Choukér
- Laboratory of Translational Research "Stress and Immunity", Department of Anesthesiology, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Marchioninistr. 15, 81377, Munich, Germany
| |
Collapse
|
21
|
Brandes F, Borrmann M, Buschmann D, Meidert AS, Reithmair M, Langkamp M, Pridzun L, Kirchner B, Billaud JN, Amin NM, Pearson JC, Klein M, Hauer D, Gevargez Zoubalan C, Lindemann A, Choukér A, Felbinger TW, Steinlein OK, Pfaffl MW, Kaufmann I, Schelling G. Progranulin signaling in sepsis, community-acquired bacterial pneumonia and COVID-19: a comparative, observational study. Intensive Care Med Exp 2021; 9:43. [PMID: 34476621 PMCID: PMC8412980 DOI: 10.1186/s40635-021-00406-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/22/2021] [Indexed: 03/28/2023] Open
Abstract
Background Progranulin is a widely expressed pleiotropic growth factor with a central regulatory effect during the early immune response in sepsis. Progranulin signaling has not been systematically studied and compared between sepsis, community-acquired pneumonia (CAP), COVID-19 pneumonia and a sterile systemic inflammatory response (SIRS). We delineated molecular networks of progranulin signaling by next-generation sequencing (NGS), determined progranulin plasma concentrations and quantified the diagnostic performance of progranulin to differentiate between the above-mentioned disorders using the established biomarkers procalcitonin (PCT), interleukin-6 (IL-6) and C-reactive protein (CRP) for comparison. Methods The diagnostic performance of progranulin was operationalized by calculating AUC and ROC statistics for progranulin and established biomarkers in 241 patients with sepsis, 182 patients with SIRS, 53 patients with CAP, 22 patients with COVID-19 pneumonia and 53 healthy volunteers. miRNAs and mRNAs in blood cells from sepsis patients (n = 7) were characterized by NGS and validated by RT-qPCR in an independent cohort (n = 39) to identify canonical gene networks associated with upregulated progranulin at sepsis onset. Results Plasma concentrations of progranulin (ELISA) in patients with sepsis were 57.5 (42.8–84.9, Q25–Q75) ng/ml and significantly higher than in CAP (38.0, 33.5–41.0 ng/ml, p < 0.001), SIRS (29.0, 25.0–35.0 ng/ml, p < 0.001) and the healthy state (28.7, 25.5–31.7 ng/ml, p < 0.001). Patients with COVID-19 had significantly higher progranulin concentrations than patients with CAP (67.6, 56.6–96.0 vs. 38.0, 33.5–41.0 ng/ml, p < 0.001). The diagnostic performance of progranulin for the differentiation between sepsis vs. SIRS (n = 423) was comparable to that of procalcitonin. AUC was 0.90 (95% CI = 0.87–0.93) for progranulin and 0.92 (CI = 0.88–0.96, p = 0.323) for procalcitonin. Progranulin showed high discriminative power to differentiate bacterial CAP from COVID-19 (sensitivity 0.91, specificity 0.94, AUC 0.91 (CI = 0.8–1.0) and performed significantly better than PCT, IL-6 and CRP. NGS and partial RT-qPCR confirmation revealed a transcriptomic network of immune cells with upregulated progranulin and sortilin transcripts as well as toll-like-receptor 4 and tumor-protein 53, regulated by miR-16 and others. Conclusions Progranulin signaling is elevated during the early antimicrobial response in sepsis and differs significantly between sepsis, CAP, COVID-19 and SIRS. This suggests that progranulin may serve as a novel indicator for the differentiation between these disorders. Trial registration: Clinicaltrials.gov registration number NCT03280576 Registered November 19, 2015. Supplementary Information The online version contains supplementary material available at 10.1186/s40635-021-00406-7.
Collapse
Affiliation(s)
- Florian Brandes
- Department of Anaesthesiology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany. .,Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University, Munich, Germany.
| | - Melanie Borrmann
- Department of Anaesthesiology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Dominik Buschmann
- Department of Anaesthesiology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany.,Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Agnes S Meidert
- Department of Anaesthesiology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Marlene Reithmair
- Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Markus Langkamp
- MEDIAGNOST Company, Aspenhausstr. 25, 72770, Reutlingen, Germany
| | - Lutz Pridzun
- MEDIAGNOST Company, Aspenhausstr. 25, 72770, Reutlingen, Germany
| | - Benedikt Kirchner
- Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | | | | | | | - Matthias Klein
- Department of Neurology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Daniela Hauer
- Department of Anaesthesiology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Clarissa Gevargez Zoubalan
- Department of Anaesthesiology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Anja Lindemann
- Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Alexander Choukér
- Department of Anaesthesiology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Thomas W Felbinger
- Department of Anaesthesiology, Neuperlach Hospital, City Hospitals of Munich, Munich, Germany
| | - Ortrud K Steinlein
- Institute of Human Genetics, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Michael W Pfaffl
- Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Ines Kaufmann
- Department of Anaesthesiology, Neuperlach Hospital, City Hospitals of Munich, Munich, Germany
| | - Gustav Schelling
- Department of Anaesthesiology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
22
|
Small RNA sequencing evaluation of renal microRNA biomarkers in dogs with X-linked hereditary nephropathy. Sci Rep 2021; 11:17437. [PMID: 34465843 PMCID: PMC8408228 DOI: 10.1038/s41598-021-96870-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/21/2021] [Indexed: 12/16/2022] Open
Abstract
Dogs with X-linked hereditary nephropathy (XLHN) are an animal model for Alport syndrome in humans and progressive chronic kidney disease (CKD). Using mRNA sequencing (mRNA-seq), we have characterized the gene expression profile affecting the progression of XLHN; however, the microRNA (miRNA, miR) expression remains unknown. With small RNA-seq and quantitative RT-PCR (qRT-PCR), we used 3 small RNA-seq analysis tools (QIAGEN OmicSoft Studio, miRDeep2, and CPSS 2.0) to profile differentially expressed renal miRNAs, top-ranked miRNA target genes, and enriched biological processes and pathways in CKD progression. Twenty-three kidney biopsies were collected from 5 dogs with XLHN and 4 age-matched, unaffected littermates at 3 clinical time points (T1: onset of proteinuria, T2: onset of azotemia, and T3: advanced azotemia). We identified up to 23 differentially expressed miRNAs at each clinical time point. Five miRNAs (miR-21, miR-146b, miR-802, miR-142, miR-147) were consistently upregulated in affected dogs. We identified miR-186 and miR-26b as effective reference miRNAs for qRT-PCR. This study applied small RNA-seq to identify differentially expressed miRNAs that might regulate critical pathways contributing to CKD progression in dogs with XLHN.
Collapse
|
23
|
Ge H, Liu G, Yamawaki TM, Tao C, Alexander ST, Ly K, Fordstrom P, Shkumatov AA, Li CM, Rajamani S, Zhou M, Ason B. Phytosterol accumulation results in ventricular arrhythmia, impaired cardiac function and death in mice. Sci Rep 2021; 11:17449. [PMID: 34465831 PMCID: PMC8408133 DOI: 10.1038/s41598-021-96936-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
Heart failure (HF) and cardiac arrhythmias share overlapping pathological mechanisms that act cooperatively to accelerate disease pathogenesis. Cardiac fibrosis is associated with both pathological conditions. Our previous work identified a link between phytosterol accumulation and cardiac injury in a mouse model of phytosterolemia, a rare disorder characterized by elevated circulating phytosterols and increased cardiovascular disease risk. Here, we uncover a previously unknown pathological link between phytosterols and cardiac arrhythmias in the same animal model. Phytosterolemia resulted in inflammatory pathway induction, premature ventricular contractions (PVC) and ventricular tachycardia (VT). Blockade of phytosterol absorption either by therapeutic inhibition or by genetic inactivation of NPC1L1 prevented the induction of inflammation and arrhythmogenesis. Inhibition of phytosterol absorption reduced inflammation and cardiac fibrosis, improved cardiac function, reduced the incidence of arrhythmias and increased survival in a mouse model of phytosterolemia. Collectively, this work identified a pathological mechanism whereby elevated phytosterols result in inflammation and cardiac fibrosis leading to impaired cardiac function, arrhythmias and sudden death. These comorbidities provide insight into the underlying pathophysiological mechanism for phytosterolemia-associated risk of sudden cardiac death.
Collapse
Affiliation(s)
- Hongfei Ge
- Cardiometabolic Disorders Therapeutic Area, Amgen Research, Amgen, Inc, 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Gongxin Liu
- Cardiometabolic Disorders Therapeutic Area, Amgen Research, Amgen, Inc, 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Tracy M Yamawaki
- Genomic Analysis Unit, Amgen Research, South San Francisco, CA, USA
| | - Caroline Tao
- Cardiometabolic Disorders Therapeutic Area, Amgen Research, Amgen, Inc, 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Shawn T Alexander
- Cardiometabolic Disorders Therapeutic Area, Amgen Research, Amgen, Inc, 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Kimberly Ly
- Translational Safety and Bioanalytical Sciences, Amgen Research, South San Francisco, CA, USA
| | - Preston Fordstrom
- Cardiometabolic Disorders Therapeutic Area, Amgen Research, Amgen, Inc, 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Artem A Shkumatov
- Translational Safety and Bioanalytical Sciences, Amgen Research, South San Francisco, CA, USA
| | - Chi-Ming Li
- Genomic Analysis Unit, Amgen Research, South San Francisco, CA, USA
| | - Sridharan Rajamani
- Translational Safety and Bioanalytical Sciences, Amgen Research, South San Francisco, CA, USA
| | - Mingyue Zhou
- Cardiometabolic Disorders Therapeutic Area, Amgen Research, Amgen, Inc, 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Brandon Ason
- Cardiometabolic Disorders Therapeutic Area, Amgen Research, Amgen, Inc, 1120 Veterans Blvd, South San Francisco, CA, 94080, USA.
| |
Collapse
|
24
|
Alser M, Rotman J, Deshpande D, Taraszka K, Shi H, Baykal PI, Yang HT, Xue V, Knyazev S, Singer BD, Balliu B, Koslicki D, Skums P, Zelikovsky A, Alkan C, Mutlu O, Mangul S. Technology dictates algorithms: recent developments in read alignment. Genome Biol 2021; 22:249. [PMID: 34446078 PMCID: PMC8390189 DOI: 10.1186/s13059-021-02443-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 07/28/2021] [Indexed: 01/08/2023] Open
Abstract
Aligning sequencing reads onto a reference is an essential step of the majority of genomic analysis pipelines. Computational algorithms for read alignment have evolved in accordance with technological advances, leading to today's diverse array of alignment methods. We provide a systematic survey of algorithmic foundations and methodologies across 107 alignment methods, for both short and long reads. We provide a rigorous experimental evaluation of 11 read aligners to demonstrate the effect of these underlying algorithms on speed and efficiency of read alignment. We discuss how general alignment algorithms have been tailored to the specific needs of various domains in biology.
Collapse
Affiliation(s)
- Mohammed Alser
- Computer Science Department, ETH Zürich, 8092, Zürich, Switzerland
- Computer Engineering Department, Bilkent University, 06800 Bilkent, Ankara, Turkey
- Information Technology and Electrical Engineering Department, ETH Zürich, Zürich, 8092, Switzerland
| | - Jeremy Rotman
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Dhrithi Deshpande
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089, USA
| | - Kodi Taraszka
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Huwenbo Shi
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Pelin Icer Baykal
- Department of Computer Science, Georgia State University, Atlanta, GA, 30302, USA
| | - Harry Taegyun Yang
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Bioinformatics Interdepartmental Ph.D. Program, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Victor Xue
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Sergey Knyazev
- Department of Computer Science, Georgia State University, Atlanta, GA, 30302, USA
| | - Benjamin D Singer
- Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Biochemistry & Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, USA
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Brunilda Balliu
- Department of Computational Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - David Koslicki
- Computer Science and Engineering, Pennsylvania State University, University Park, PA, 16801, USA
- Biology Department, Pennsylvania State University, University Park, PA, 16801, USA
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16801, USA
| | - Pavel Skums
- Department of Computer Science, Georgia State University, Atlanta, GA, 30302, USA
| | - Alex Zelikovsky
- Department of Computer Science, Georgia State University, Atlanta, GA, 30302, USA
- The Laboratory of Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Can Alkan
- Computer Engineering Department, Bilkent University, 06800 Bilkent, Ankara, Turkey
- Bilkent-Hacettepe Health Sciences and Technologies Program, Ankara, Turkey
| | - Onur Mutlu
- Computer Science Department, ETH Zürich, 8092, Zürich, Switzerland
- Computer Engineering Department, Bilkent University, 06800 Bilkent, Ankara, Turkey
- Information Technology and Electrical Engineering Department, ETH Zürich, Zürich, 8092, Switzerland
| | - Serghei Mangul
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
25
|
Antitumor Potency of an Anti-CD19 Chimeric Antigen Receptor T-Cell Therapy, Lisocabtagene Maraleucel in Combination With Ibrutinib or Acalabrutinib. J Immunother 2021; 43:107-120. [PMID: 31899702 PMCID: PMC7144752 DOI: 10.1097/cji.0000000000000307] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Supplemental Digital Content is available in the text. Chimeric antigen receptor (CAR) T-cell therapy is a promising treatment for patients with CD19+ B-cell malignancies. Combination strategies that improve CAR T-cell potency, limit tumor environment–mediated immune dysfunction, and directly reduce tumor burden may increase the potential for durable clinical benefit of CAR T-cell therapy. Lisocabtagene maraleucel (liso-cel) is a product therapy candidate being tested in patients with relapsed/refractory non-Hodgkin lymphoma or chronic lymphocytic leukemia. This study assessed the in vitro and in vivo functionality of CAR T cells transduced to express the anti-CD19 CAR of liso-cel in combination with ibrutinib or acalabrutinib. In prolonged stimulation assays, the presence of ibrutinib or acalabrutinib improved the CAR T-cell effector function. RNA-Seq analysis and surface marker profiling of these CAR T cells treated with ibrutinib but not acalabrutinib revealed gene expression changes consistent with skewing toward a memory-like, type 1 T-helper, Bruton tyrosine kinase phenotype. Ibrutinib or acalabrutinib improved CD19+ tumor clearance and prolonged survival of tumor-bearing mice when used in combination with CAR T cells. A combination of the defined cell product therapy candidate, liso-cel, with ibrutinib or acalabrutinib is an attractive approach that may potentiate the promising clinical responses already achieved in CD19+ B-cell malignancies with each of these single agents.
Collapse
|
26
|
Decato BE, Ammar R, Reinke-Breen L, Thompson JR, Azzara AV. Transcriptome analysis reveals key genes modulated by ALK5 inhibition in a bleomycin model of systemic sclerosis. Rheumatology (Oxford) 2021; 61:1717-1727. [PMID: 34289031 PMCID: PMC8996787 DOI: 10.1093/rheumatology/keab580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 07/09/2021] [Indexed: 11/14/2022] Open
Abstract
Objective SSc is a rheumatic autoimmune disease affecting roughly 20 000 people worldwide and characterized by excessive collagen accumulation in the skin and internal organs. Despite the high morbidity and mortality associated with SSc, there are no approved disease-modifying agents. Our objective in this study was to explore transcriptomic and model-based drug discovery approaches for SSc. Methods In this study, we explored the molecular basis for SSc pathogenesis in a well-studied mouse model of scleroderma. We profiled the skin and lung transcriptomes of mice at multiple timepoints, analysing the differential gene expression that underscores the development and resolution of bleomycin-induced fibrosis. Results We observed shared expression signatures of upregulation and downregulation in fibrotic skin and lung tissue, and observed significant upregulation of key pro-fibrotic genes including GDF15, Saa3, Cxcl10, Spp1 and Timp1. To identify changes in gene expression in responses to anti-fibrotic therapy, we assessed the effect of TGF-β pathway inhibition via oral ALK5 (TGF-β receptor I) inhibitor SB525334 and observed a time-lagged response in the lung relative to skin. We also implemented a machine learning algorithm that showed promise at predicting lung function using transcriptome data from both skin and lung biopsies. Conclusion This study provides the most comprehensive look at the gene expression dynamics of an animal model of SSc to date, provides a rich dataset for future comparative fibrotic disease research, and helps refine our understanding of pathways at work during SSc pathogenesis and intervention.
Collapse
Affiliation(s)
- Benjamin E Decato
- Research & Early Development, Bristol-Myers Squibb Company, Route 206 & Province Line Rd, Lawrenceville, NJ, 08543, USA
| | - Ron Ammar
- Research & Early Development, Bristol-Myers Squibb Company, Route 206 & Province Line Rd, Lawrenceville, NJ, 08543, USA
| | - Lauren Reinke-Breen
- Research & Early Development, Bristol-Myers Squibb Company, Route 206 & Province Line Rd, Lawrenceville, NJ, 08543, USA
| | - John R Thompson
- Research & Early Development, Bristol-Myers Squibb Company, Route 206 & Province Line Rd, Lawrenceville, NJ, 08543, USA
| | - Anthony V Azzara
- Research & Early Development, Bristol-Myers Squibb Company, Route 206 & Province Line Rd, Lawrenceville, NJ, 08543, USA
| |
Collapse
|
27
|
Ningappa M, Adenuga M, Ngo KA, Mohamed N, Narayanan T, Prasadan K, Ashokkumar C, Das J, Schmitt L, Hartman H, Sehrawat A, Salgado CM, Reyes-Mugica M, Gittes GK, Lo CW, Subramaniam S, Sindhi R. Mechanisms of Impaired Lung Development and Ciliation in Mannosidase-1-Alpha-2 ( Man1a2) Mutants. Front Physiol 2021; 12:658518. [PMID: 34366878 PMCID: PMC8343402 DOI: 10.3389/fphys.2021.658518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/03/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Ciliary defects cause heterogenous phenotypes related to mutation burden which lead to impaired development. A previously reported homozygous deletion in the Man1a2 gene causes lethal respiratory failure in newborn pups and decreased lung ciliation compared with wild type (WT) pups. The effects of heterozygous mutation, and the potential for rescue are not known. PURPOSE We hypothesized that survival and lung ciliation, (a) would decrease progressively in Man1a2 +/- heterozygous and Man1a2 -/- null newborn pups compared with WT, and (b) could be enhanced by gestational treatment with N-Acetyl-cysteine (NAC), an antioxidant. METHODS Man1a2+/- adult mice were fed NAC or placebo from a week before breeding through gestation. Survival of newborn pups was monitored for 24 h. Lungs, liver and tails were harvested for morphology, genotyping, and transcriptional profiling. RESULTS Survival (p = 0.0001, Kaplan-Meier) and percent lung ciliation (p = 0.0001, ANOVA) measured by frequency of Arl13b+ respiratory epithelial cells decreased progressively, as hypothesized. Compared with placebo, gestational NAC treatment enhanced (a) lung ciliation in pups with each genotype, (b) survival in heterozygous pups (p = 0.017) but not in WT or null pups. Whole transcriptome of lung but not liver demonstrated patterns of up- and down-regulated genes that were identical in living heterozygous and WT pups, and completely opposite to those in dead heterozygous and null pups. Systems biology analysis enabled reconstruction of protein interaction networks that yielded functionally relevant modules and their interactions. In these networks, the mutant Man1a2 enzyme contributes to abnormal synthesis of proteins essential for lung development. The associated unfolded protein, hypoxic and oxidative stress responses can be mitigated with NAC. Comparisons with the developing human fetal lung transcriptome show that NAC likely restores normal vascular and epithelial tube morphogenesis in Man1a2 mutant mice. CONCLUSION Survival and lung ciliation in the Man1a2 mutant mouse, and its improvement with N-Acetyl cysteine is genotype-dependent. NAC-mediated rescue depends on the central role for oxidative and hypoxic stress in regulating ciliary function and organogenesis during development.
Collapse
Affiliation(s)
- Mylarappa Ningappa
- Hillman Center for Pediatric Transplantation, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Morayooluwa Adenuga
- Hillman Center for Pediatric Transplantation, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Kim A. Ngo
- Department of Bioengineering, University of California, San Diego, San Diego, La Jolla, CA, United States
| | - Nada Mohamed
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Tejaswini Narayanan
- Department of Bioengineering, University of California, San Diego, San Diego, La Jolla, CA, United States
| | - Krishna Prasadan
- Rangos Research Center Animal Imaging Core, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Chethan Ashokkumar
- Hillman Center for Pediatric Transplantation, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Jishnu Das
- Hillman Center for Pediatric Transplantation, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
- Departments of Immunology and Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lori Schmitt
- Histology Core Laboratory Manager, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Hannah Hartman
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Anuradha Sehrawat
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Claudia M. Salgado
- Division of Pediatric Pathology, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Miguel Reyes-Mugica
- Division of Pediatric Pathology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - George K. Gittes
- Surgeon-in-Chief Emeritus, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Cecilia W. Lo
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, San Diego, La Jolla, CA, United States
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, La Jolla, CA, United States
- Department of Computer Science and Engineering, and Nanoengineering, University of California, San Diego, San Diego, La Jolla, CA, United States
| | - Rakesh Sindhi
- Hillman Center for Pediatric Transplantation, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| |
Collapse
|
28
|
Wigger L, Barovic M, Brunner AD, Marzetta F, Schöniger E, Mehl F, Kipke N, Friedland D, Burdet F, Kessler C, Lesche M, Thorens B, Bonifacio E, Legido-Quigley C, Barbier Saint Hilaire P, Delerive P, Dahl A, Klose C, Gerl MJ, Simons K, Aust D, Weitz J, Distler M, Schulte AM, Mann M, Ibberson M, Solimena M. Multi-omics profiling of living human pancreatic islet donors reveals heterogeneous beta cell trajectories towards type 2 diabetes. Nat Metab 2021; 3:1017-1031. [PMID: 34183850 DOI: 10.1038/s42255-021-00420-9] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/21/2021] [Indexed: 12/19/2022]
Abstract
Most research on human pancreatic islets is conducted on samples obtained from normoglycaemic or diseased brain-dead donors and thus cannot accurately describe the molecular changes of pancreatic islet beta cells as they progress towards a state of deficient insulin secretion in type 2 diabetes (T2D). Here, we conduct a comprehensive multi-omics analysis of pancreatic islets obtained from metabolically profiled pancreatectomized living human donors stratified along the glycemic continuum, from normoglycemia to T2D. We find that islet pools isolated from surgical samples by laser-capture microdissection display remarkably more heterogeneous transcriptomic and proteomic profiles in patients with diabetes than in non-diabetic controls. The differential regulation of islet gene expression is already observed in prediabetic individuals with impaired glucose tolerance. Our findings demonstrate a progressive, but disharmonic, remodelling of mature beta cells, challenging current hypotheses of linear trajectories toward precursor or transdifferentiation stages in T2D. Furthermore, through integration of islet transcriptomics with preoperative blood plasma lipidomics, we define the relative importance of gene coexpression modules and lipids that are positively or negatively associated with HbA1c levels, pointing to potential prognostic markers.
Collapse
Affiliation(s)
- Leonore Wigger
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Marko Barovic
- Department of Molecular Diabetology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | | | - Flavia Marzetta
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Eyke Schöniger
- Department of Molecular Diabetology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Florence Mehl
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Nicole Kipke
- Department of Molecular Diabetology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Daniela Friedland
- Department of Molecular Diabetology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Frederic Burdet
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Camille Kessler
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Mathias Lesche
- DRESDEN-concept Genome Center, c/o Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Ezio Bonifacio
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Center for Regenerative Therapies Dresden, Faculty of Medicine and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | | | | | - Philippe Delerive
- Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France
| | - Andreas Dahl
- DRESDEN-concept Genome Center, c/o Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | | | | | | | - Daniela Aust
- Department of Pathology, Medical Faculty, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- NCT Biobank Dresden, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Marius Distler
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Anke M Schulte
- Sanofi-Aventis Deutschland GmbH, Diabetes Research, Industriepark Höchst, Frankfurt am Main, Germany
| | - Matthias Mann
- Max Planck Institute of Biochemistry, Martinsried, Germany.
| | - Mark Ibberson
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Michele Solimena
- Department of Molecular Diabetology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany.
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.
| |
Collapse
|
29
|
Luo X, Yin J, Dwyer D, Yamawaki T, Zhou H, Ge H, Han CY, Shkumatov A, Snyder K, Ason B, Li CM, Homann O, Stolina M. Chamber-enriched gene expression profiles in failing human hearts with reduced ejection fraction. Sci Rep 2021; 11:11839. [PMID: 34088950 PMCID: PMC8178406 DOI: 10.1038/s41598-021-91214-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
Heart failure with reduced ejection fraction (HFrEF) constitutes 50% of HF hospitalizations and is characterized by high rates of mortality. To explore the underlying mechanisms of HFrEF etiology and progression, we studied the molecular and cellular differences in four chambers of non-failing (NF, n = 10) and HFrEF (n = 12) human hearts. We identified 333 genes enriched within NF heart subregions and often associated with cardiovascular disease GWAS variants. Expression analysis of HFrEF tissues revealed extensive disease-associated transcriptional and signaling alterations in left atrium (LA) and left ventricle (LV). Common left heart HFrEF pathologies included mitochondrial dysfunction, cardiac hypertrophy and fibrosis. Oxidative stress and cardiac necrosis pathways were prominent within LV, whereas TGF-beta signaling was evident within LA. Cell type composition was estimated by deconvolution and revealed that HFrEF samples had smaller percentage of cardiomyocytes within the left heart, higher representation of fibroblasts within LA and perivascular cells within the left heart relative to NF samples. We identified essential modules associated with HFrEF pathology and linked transcriptome discoveries with human genetics findings. This study contributes to a growing body of knowledge describing chamber-specific transcriptomics and revealed genes and pathways that are associated with heart failure pathophysiology, which may aid in therapeutic target discovery.
Collapse
Affiliation(s)
- Xin Luo
- Genome Analysis Unit, Amgen Research, 1120 Veterans BLVD, South San Francisco, CA, 94010, USA
| | - Jun Yin
- Genome Analysis Unit, Amgen Research, 1120 Veterans BLVD, South San Francisco, CA, 94010, USA
| | - Denise Dwyer
- Department of Cardiometabolic Disorders, Amgen Research, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Tracy Yamawaki
- Genome Analysis Unit, Amgen Research, 1120 Veterans BLVD, South San Francisco, CA, 94010, USA
| | - Hong Zhou
- Genome Analysis Unit, Amgen Research, 1120 Veterans BLVD, South San Francisco, CA, 94010, USA
| | - Hongfei Ge
- Department of Cardiometabolic Disorders, Amgen Research, 1120 Veterans BLVD, South San Francisco, CA, 94010, USA
| | - Chun-Ya Han
- Department of Cardiometabolic Disorders, Amgen Research, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Artem Shkumatov
- TS&BA Pathology, Amgen Research, 1120 Veterans BLVD, South San Francisco, CA, 94010, USA
| | - Karen Snyder
- Clinical Biomarkers, Amgen Research, 1120 Veterans BLVD, South San Francisco, CA, 94010, USA
| | - Brandon Ason
- Department of Cardiometabolic Disorders, Amgen Research, 1120 Veterans BLVD, South San Francisco, CA, 94010, USA
| | - Chi-Ming Li
- Genome Analysis Unit, Amgen Research, 1120 Veterans BLVD, South San Francisco, CA, 94010, USA
| | - Oliver Homann
- Genome Analysis Unit, Amgen Research, 1120 Veterans BLVD, South San Francisco, CA, 94010, USA
| | - Marina Stolina
- Department of Cardiometabolic Disorders, Amgen Research, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| |
Collapse
|
30
|
Veit JGS, Poumay Y, Mendes D, Kreitinger J, Walker L, Paquet A, Menigot C, Zolezzi F, Paller AS, Diaz P. Preclinical assessment of dual CYP26[A1/B1] inhibitor, DX308, as an improved treatment for keratinization disorders. SKIN HEALTH AND DISEASE 2021; 1:e22. [PMID: 35664983 PMCID: PMC9060145 DOI: 10.1002/ski2.22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 11/30/2022]
Abstract
Background Retinoid‐based therapies are commonly used in the treatment of disorders of keratinization and other skin disorders but can result in non‐specific effects and adverse reactions. Use of retinoic acid metabolism blocking agents (RAMBAs) such as DX308 may address these shortcomings. Objectives Characterize the therapeutic potential of recently discovered, CYP26‐selective RAMBA, DX308. Materials and Methods Preliminary in vitro assessment of potential off‐target activity, metabolic and toxicologic profiling. Studies to assess safety and efficacy of topical treatment in correcting abnormal skin morphology in rhino mice. Extensive gene expression profiling by RNA sequencing and qPCR in 3D epidermis grown with keratinocytes (KCs) from keratinization disorders and healthy controls, to investigate modulation of retinoid biopathways. Results In vitro, DX308 does not interact with off‐target nuclear receptors or CYP450s, is not genotoxic, and is stable in skin, despite vigorous hepatic metabolism. In vivo, topical DX308 induces comedolysis and epidermal thickening without apparent adverse effects. Gene expression profiling shows potent modulation of retinoid‐responsive genes by DX308 in both healthy and keratinization disorder KCs. Pathway analysis suggests DX308 may inhibit inflammatory and immune responses in KCs. Conclusions These preliminary studies suggest that DX308 is an efficacious topical therapeutic with a favourable metabolic and safety profiles. DX308 may present an improved therapeutic alternative for the treatment of keratinization disorders and other retinoid‐responsive skin ailments.
Collapse
Affiliation(s)
- J G S Veit
- Department of Biomedical and Pharmaceutical Sciences University of Montana Missoula Montana USA.,URPHYM-NARILIS University of Namur Namur Belgium.,R&D DermaXon LLC Missoula Montana USA
| | - Y Poumay
- URPHYM-NARILIS University of Namur Namur Belgium
| | - D Mendes
- R&D DermaXon LLC Missoula Montana USA
| | | | - L Walker
- R&D DermaXon LLC Missoula Montana USA
| | | | | | | | - A S Paller
- Department of Dermatology Northwestern University Feinberg School of Medicine Chicago Illinois USA
| | - P Diaz
- Department of Biomedical and Pharmaceutical Sciences University of Montana Missoula Montana USA.,R&D DermaXon LLC Missoula Montana USA
| |
Collapse
|
31
|
Tran JQ, Grover D, Zhang M, Stapels M, Brennan R, Bangari DS, Piepenhagen PA, Roberts E, Oliva P, Zubair F, Vela JL, Richards SM, Joseph AM. Expansion of immature, nucleated red blood cells by transient low-dose methotrexate immune tolerance induction in mice. Clin Exp Immunol 2021; 203:409-423. [PMID: 33205401 PMCID: PMC7874831 DOI: 10.1111/cei.13552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 11/28/2022] Open
Abstract
Biological treatments such as enzyme-replacement therapies (ERT) can generate anti-drug antibodies (ADA), which may reduce drug efficacy and impact patient safety and consequently led to research to mitigate ADA responses. Transient low-dose methotrexate (TLD-MTX) as a prophylactic ITI regimen, when administered concurrently with ERT, induces long-lived reduction of ADA to recombinant human alglucosidase alfa (rhGAA) in mice. In current clinical practice, a prophylactic ITI protocol that includes TLD-MTX, rituximab and intravenous immunoglobulin (optional), successfully induced lasting control of ADA to rhGAA in high-risk, cross-reactive immunological material (CRIM)-negative infantile-onset Pompe disease (IOPD) patients. More recently, evaluation of TLD-MTX demonstrated benefit in CRIM-positive IOPD patients. To more clearly understand the mechanism for the effectiveness of TLD-MTX, non-targeted transcriptional and proteomic screens were conducted and revealed up-regulation of erythropoiesis signatures. Confirmatory studies showed transiently larger spleens by weight, increased spleen cellularity and that following an initial reduction of mature red blood cells (RBCs) in the bone marrow and blood, a significant expansion of Ter-119+ CD71+ immature RBCs was observed in spleen and blood of mice. Histology sections revealed increased nucleated cells, including hematopoietic precursors, in the splenic red pulp of these mice. This study demonstrated that TLD-MTX induced a transient reduction of mature RBCs in the blood and immature RBCs in the bone marrow followed by significant enrichment of immature, nucleated RBCs in the spleen and blood during the time of immune tolerance induction, which suggested modulation of erythropoiesis may be associated with the induction of immune tolerance to rhGAA.
Collapse
Affiliation(s)
- J. Q. Tran
- Sanofi Immunology and Inflammation Research Therapeutic AreaCambridgeMAUSA
| | - D. Grover
- Sanofi Immunology and Inflammation Research Therapeutic AreaCambridgeMAUSA
| | - M. Zhang
- Sanofi Translational Sciences BioinformaticsCambridgeMAUSA
| | - M. Stapels
- Sanofi Biologics DevelopmentCambridgeMAUSA
| | | | | | | | - E. Roberts
- Sanofi Translational In Vivo ModelsCambridgeMAUSA
| | - P. Oliva
- Sanofi Immunology and Inflammation Research Therapeutic AreaCambridgeMAUSA
| | - F. Zubair
- Sanofi Immunology and Inflammation Research Therapeutic AreaCambridgeMAUSA
| | - J. L. Vela
- Sanofi Immunology and Inflammation Research Therapeutic AreaCambridgeMAUSA
| | - S. M. Richards
- Sanofi Translational Medicine and Early DevelopmentCambridgeMAUSA
| | - A. M. Joseph
- Sanofi Immunology and Inflammation Research Therapeutic AreaCambridgeMAUSA
| |
Collapse
|
32
|
Simoneau J, Gosselin R, Scott MS. Factorial study of the RNA-seq computational workflow identifies biases as technical gene signatures. NAR Genom Bioinform 2021; 2:lqaa043. [PMID: 33575596 PMCID: PMC7671328 DOI: 10.1093/nargab/lqaa043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/15/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
RNA-seq is a modular experimental and computational approach aiming in identifying and quantifying RNA molecules. The modularity of the RNA-seq technology enables adaptation of the protocol to develop new ways to explore RNA biology, but this modularity also brings forth the importance of methodological thoroughness. Liberty of approach comes with the responsibility of choices, and such choices must be informed. Here, we present an approach that identifies gene group-specific quantification biases in current RNA-seq software and references by processing datasets using diverse RNA-seq computational pipelines, and by decomposing these expression datasets with an independent component analysis matrix factorization method. By exploring the RNA-seq pipeline using this systemic approach, we identify genome annotations as a design choice that affects to the same extent quantification results as does the choice of aligners and quantifiers. We also show that the different choices in RNA-seq methodology are not independent, identifying interactions between genome annotations and quantification software. Genes were mainly affected by differences in their sequence, by overlapping genes and genes with similar sequence. Our approach offers an explanation for the observed biases by identifying the common features used differently by the software and references, therefore providing leads for the betterment of RNA-seq methodology.
Collapse
Affiliation(s)
- Joël Simoneau
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, J1K 2R1, Canada
| | - Ryan Gosselin
- Department of Chemical & Biotechnological Engineering, Faculty of Engineering, Université de Sherbrooke, Sherbrooke, Québec, J1K 2R1, Canada
| | - Michelle S Scott
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, J1K 2R1, Canada
| |
Collapse
|
33
|
Castañeda TR, Méndez M, Davison I, Elvert R, Schwahn U, Boldina G, Rocher C, Scherer P, Singh K, Bangari DS, Falkenhahn M, Kannt A, Konkar A, Larsen PJ, Arbeeny C, Dhal PK, Hübschle T. The Novel Phosphate and Bile Acid Sequestrant Polymer SAR442357 Delays Disease Progression in a Rat Model of Diabetic Nephropathy. J Pharmacol Exp Ther 2021; 376:190-203. [PMID: 33203659 DOI: 10.1124/jpet.120.000285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
As a gut-restricted, nonabsorbed therapy, polymeric bile acid sequestrants (BAS) play an important role in managing hyperlipidemia and hyperglycemia. Similarly, nonabsorbable sequestrants of dietary phosphate have been used for the management of hyperphosphatemia in end-stage renal disease. To evaluate the potential utility of such polymer sequestrants to treat type 2 diabetes (T2D) and its associated renal and cardiovascular complications, we synthesized a novel polymeric sequestrant, SAR442357, possessing optimized bile acid (BA) and phosphate sequestration characteristics. Long-term treatment of T2D obese cZucker fatty/Spontaneously hypertensive heart failure F1 hybrid (ZSF1) with SAR442357 resulted in enhanced sequestration of BAs and phosphate in the gut, improved glycemic control, lowering of serum cholesterol, and attenuation of diabetic kidney disease (DKD) progression. In comparison, colesevelam, a BAS with poor phosphate binding properties, did not prevent DKD progression, whereas losartan, an angiotensin II receptor blocker that is widely used to treat DKD, showed no effect on hyperglycemia. Analysis of hepatic gene expression levels of the animals treated with SAR442357 revealed upregulation of genes responsible for the biosynthesis of cholesterol and BAs, providing clear evidence of target engagement and mode of action of the new sequestrant. Additional hepatic gene expression pathway changes were indicative of an interruption of the enterohepatic BA cycle. Histopathological analysis of ZSF1 rat kidneys treated with SAR442357 further supported its nephroprotective properties. Collectively, these findings reveal the pharmacological benefit of simultaneous sequestration of BAs and phosphate in treating T2D and its associated comorbidities and cardiovascular complications. SIGNIFICANCE STATEMENT: A new nonabsorbed polymeric sequestrant with optimum phosphate and bile salt sequestration properties was developed as a treatment option for DKD. The new polymeric sequestrant offered combined pharmacological benefits including glucose regulation, lipid lowering, and attenuation of DKD progression in a single therapeutic agent.
Collapse
Affiliation(s)
- Tamara R Castañeda
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - María Méndez
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Ian Davison
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Ralf Elvert
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Uwe Schwahn
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Galina Boldina
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Corinne Rocher
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Petra Scherer
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Kuldeep Singh
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Dinesh S Bangari
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Mechthilde Falkenhahn
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Aimo Kannt
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Anish Konkar
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Philip J Larsen
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Cynthia Arbeeny
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Pradeep K Dhal
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| | - Thomas Hübschle
- R&D Diabetes (T.R.C., R.E., A.Ka., A.Ko., P.J.L., C.A., T.H.), Integrated Drug Discovery (M.M.), Biomarkers and Clinical Bioanalysis (U.S.), Translational In Vivo Models, Global Discovery Pathology (P.S.), and Global Research Project Management (M.F.), Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany; C&BD Haverhill Operations, Sanofi GB Genzyme Limited, Haverhill, Suffolk, United Kingdom (I.D.); R&D Translational Sciences France, Bioinformatics, Sanofi, Chilly-Mazarin Cedex, France (C.R.); Translational In Vivo Models, Global Discovery Pathology, Framingham, Massachusetts (K.S., D.S.B.); and Pharmaceutical Development Platform, Sanofi Global R&D, Waltham, Massachusetts (P.K.D.)
| |
Collapse
|
34
|
Duriez M, Jacquet A, Hoet L, Roche S, Bock MD, Rocher C, Haussy G, Vigé X, Bocskei Z, Slavnic T, Martin V, Guillemot JC, Didier M, Kannt A, Orsini C, Mikol V, Fèvre ACL. A 3D Human Liver Model of Nonalcoholic Steatohepatitis. J Clin Transl Hepatol 2020; 8:359-370. [PMID: 33447518 PMCID: PMC7782122 DOI: 10.14218/jcth.2020.00015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/19/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022] Open
Abstract
Background and Aims: To better understand nonalcoholic steatohepatitis (NASH) disease progression and to evaluate drug targets and compound activity, we undertook the development of an in vitro 3D model to mimic liver architecture and the NASH environment. Methods: We have developed an in vitro preclinical 3D NASH model by coculturing primary human hepatocytes, human stellate cells, liver endothelial cells and Kupffer cells embedded in a hydrogel of rat collagen on a 96-well plate. A NASH-like environment was induced by addition of medium containing free fatty acids and tumor necrosis factor-α. This model was then characterized by biochemical, imaging and transcriptomics analyses. Results: We succeeded in defining suitable culture conditions to maintain the 3D coculture for up to 10 days in vitro, with the lowest level of steatosis and reproducible low level of inflammation and fibrosis. NASH disease was induced with a custom medium mimicking NASH features. The cell model exhibited the key NASH disease phenotypes of hepatocyte injury, steatosis, inflammation, and fibrosis. Hepatocyte injury was highlighted by a decrease of CYP3A4 expression and activity, without loss of viability up to day 10. Moreover, the model was able to stimulate a stable inflammatory and early fibrotic environment, with expression and secretion of several cytokines. A global gene expression analysis confirmed the NASH induction. Conclusions: This is a new in vitro model of NASH disease consisting of four human primary cell-types that exhibits most features of the disease. The 10-day cell viability and cost effectiveness of the model make it suitable for medium throughput drug screening and provide attractive avenues to better understand disease physiology and to identify and characterize new drug targets.
Collapse
Affiliation(s)
- Marion Duriez
- Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin, France
| | - Agnes Jacquet
- Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin, France
| | - Lucile Hoet
- Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin, France
| | - Sandrine Roche
- Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin, France
| | - Marie-Dominique Bock
- Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin, France
| | - Corinne Rocher
- Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin, France
| | - Gilles Haussy
- Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin, France
| | - Xavier Vigé
- Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin, France
| | - Zsolt Bocskei
- Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin, France
| | | | - Valérie Martin
- Non Clinical Biostatistics, Sanofi, Vitry sur Seine, France
| | | | - Michel Didier
- Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin, France
| | - Aimo Kannt
- Diabetes and Cardiovascular Research, Sanofi, Industriepark Höchst, Frankfurt, Germany
- Present address: Institute of Experimental Pharmacology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Cécile Orsini
- Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin, France
| | - Vincent Mikol
- Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin, France
| | - Anne-Céline Le Fèvre
- Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, Chilly-Mazarin, France
- Correspondence to: Anne-Céline Le Fèvre, Translational Sciences, Sanofi, 1 Avenue Pierre Brossolette, 91385 Chilly-Mazarin, France. Tel: +33-160495815, E-mail:
| |
Collapse
|
35
|
Wisdom AJ, Mowery YM, Hong CS, Himes JE, Nabet BY, Qin X, Zhang D, Chen L, Fradin H, Patel R, Bassil AM, Muise ES, King DA, Xu ES, Carpenter DJ, Kent CL, Smythe KS, Williams NT, Luo L, Ma Y, Alizadeh AA, Owzar K, Diehn M, Bradley T, Kirsch DG. Single cell analysis reveals distinct immune landscapes in transplant and primary sarcomas that determine response or resistance to immunotherapy. Nat Commun 2020; 11:6410. [PMID: 33335088 PMCID: PMC7746723 DOI: 10.1038/s41467-020-19917-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy fails to cure most cancer patients. Preclinical studies indicate that radiotherapy synergizes with immunotherapy, promoting radiation-induced antitumor immunity. Most preclinical immunotherapy studies utilize transplant tumor models, which overestimate patient responses. Here, we show that transplant sarcomas are cured by PD-1 blockade and radiotherapy, but identical treatment fails in autochthonous sarcomas, which demonstrate immunoediting, decreased neoantigen expression, and tumor-specific immune tolerance. We characterize tumor-infiltrating immune cells from transplant and primary tumors, revealing striking differences in their immune landscapes. Although radiotherapy remodels myeloid cells in both models, only transplant tumors are enriched for activated CD8+ T cells. The immune microenvironment of primary murine sarcomas resembles most human sarcomas, while transplant sarcomas resemble the most inflamed human sarcomas. These results identify distinct microenvironments in murine sarcomas that coevolve with the immune system and suggest that patients with a sarcoma immune phenotype similar to transplant tumors may benefit most from PD-1 blockade and radiotherapy.
Collapse
Affiliation(s)
- Amy J Wisdom
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Yvonne M Mowery
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA.
- Duke Cancer Institute, Durham, NC, 27708, USA.
| | - Cierra S Hong
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Jonathon E Himes
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Barzin Y Nabet
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Department of Oncology Biomarker Development, Genentech, South San Francisco, CA, 94080, USA
| | - Xiaodi Qin
- Duke Cancer Institute, Durham, NC, 27708, USA
| | | | - Lan Chen
- Merck & Co., Inc, Kenilworth, NJ, 07033, USA
| | - Hélène Fradin
- Duke Center for Genomic and Computational Biology, Durham, NC, 27708, USA
| | - Rutulkumar Patel
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Alex M Bassil
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | | | - Daniel A King
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Eric S Xu
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - David J Carpenter
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Collin L Kent
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | | | - Nerissa T Williams
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Ash A Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Kouros Owzar
- Duke Cancer Institute, Durham, NC, 27708, USA
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, 27710, USA
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, 94305, USA
| | - Todd Bradley
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Pediatric Genomic Medicine, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - David G Kirsch
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA.
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA.
- Duke Cancer Institute, Durham, NC, 27708, USA.
| |
Collapse
|
36
|
Tong L, Wu PY, Phan JH, Hassazadeh HR, Tong W, Wang MD. Impact of RNA-seq data analysis algorithms on gene expression estimation and downstream prediction. Sci Rep 2020; 10:17925. [PMID: 33087762 PMCID: PMC7578822 DOI: 10.1038/s41598-020-74567-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 08/27/2020] [Indexed: 11/23/2022] Open
Abstract
To use next-generation sequencing technology such as RNA-seq for medical and health applications, choosing proper analysis methods for biomarker identification remains a critical challenge for most users. The US Food and Drug Administration (FDA) has led the Sequencing Quality Control (SEQC) project to conduct a comprehensive investigation of 278 representative RNA-seq data analysis pipelines consisting of 13 sequence mapping, three quantification, and seven normalization methods. In this article, we focused on the impact of the joint effects of RNA-seq pipelines on gene expression estimation as well as the downstream prediction of disease outcomes. First, we developed and applied three metrics (i.e., accuracy, precision, and reliability) to quantitatively evaluate each pipeline's performance on gene expression estimation. We then investigated the correlation between the proposed metrics and the downstream prediction performance using two real-world cancer datasets (i.e., SEQC neuroblastoma dataset and the NIH/NCI TCGA lung adenocarcinoma dataset). We found that RNA-seq pipeline components jointly and significantly impacted the accuracy of gene expression estimation, and its impact was extended to the downstream prediction of these cancer outcomes. Specifically, RNA-seq pipelines that produced more accurate, precise, and reliable gene expression estimation tended to perform better in the prediction of disease outcome. In the end, we provided scenarios as guidelines for users to use these three metrics to select sensible RNA-seq pipelines for the improved accuracy, precision, and reliability of gene expression estimation, which lead to the improved downstream gene expression-based prediction of disease outcome.
Collapse
Affiliation(s)
- Li Tong
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Po-Yen Wu
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - John H Phan
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Hamid R Hassazadeh
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Weida Tong
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - May D Wang
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
37
|
Non-Human Primate Blood-Brain Barrier and In Vitro Brain Endothelium: From Transcriptome to the Establishment of a New Model. Pharmaceutics 2020; 12:pharmaceutics12100967. [PMID: 33066641 PMCID: PMC7602447 DOI: 10.3390/pharmaceutics12100967] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/02/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
The non-human primate (NHP)-brain endothelium constitutes an essential alternative to human in the prediction of molecule trafficking across the blood–brain barrier (BBB). This study presents a comparison between the NHP transcriptome of freshly isolated brain microcapillaries and in vitro-selected brain endothelial cells (BECs), focusing on important BBB features, namely tight junctions, receptors mediating transcytosis (RMT), ABC and SLC transporters, given its relevance as an alternative model for the molecule trafficking prediction across the BBB and identification of new brain-specific transport mechanisms. In vitro BECs conserved most of the BBB key elements for barrier integrity and control of molecular trafficking. The function of RMT via the transferrin receptor (TFRC) was characterized in this NHP-BBB model, where both human transferrin and anti-hTFRC antibody showed increased apical-to-basolateral passage in comparison to control molecules. In parallel, eventual BBB-related regional differences were Investig.igated in seven-day in vitro-selected BECs from five brain structures: brainstem, cerebellum, cortex, hippocampus, and striatum. Our analysis retrieved few differences in the brain endothelium across brain regions, suggesting a rather homogeneous BBB function across the brain parenchyma. The presently established NHP-derived BBB model closely mimics the physiological BBB, thus representing a ready-to-use tool for assessment of the penetration of biotherapeutics into the human CNS.
Collapse
|
38
|
Dash AB, Zhang J, Shen L, Li B, Berg D, Lin J, Avet‐Loiseau H, Bahlis NJ, Moreau P, Richardson PG, Di Bacco A. Clinical benefit of ixazomib plus lenalidomide-dexamethasone in myeloma patients with non-canonical NF-κB pathway activation. Eur J Haematol 2020; 105:274-285. [PMID: 32350909 PMCID: PMC7497066 DOI: 10.1111/ejh.13435] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/23/2020] [Accepted: 04/27/2020] [Indexed: 01/27/2023]
Abstract
OBJECTIVES Evaluating potential relationships between progression-free survival (PFS) and tumor gene expression patterns and mutational status was an exploratory objective of the phase 3 TOURMALINE-MM1 study (NCT01564537) of ixazomib-lenalidomide-dexamethasone (IRd) vs placebo-Rd in 722 patients with relapsed/refractory multiple myeloma (MM). METHODS We utilized gene expression and mutation data from screening bone marrow aspirates to identify tumors with non-canonical nuclear factor-κB (NF-κB) signaling pathway activation. RESULTS DNA/RNA sequencing data were available for 339 (47.0%)/399 (55.2%) patients; 49/339 (14.5%) patients had non-canonical NF-κB pathway gene mutations (tumor-necrosis-factor receptor-associated factor 2, 3 [TRAF2, TRAF3], baculoviral-inhibitor-of-apoptosis repeat-containing 2/3 [BIRC2/3]), and PFS was significantly longer with IRd vs placebo-Rd in these patients (hazard ratio [HR] 0.23). In patients with lower TRAF3 expression (median not reached vs 11 months, HR 0.47) and higher NF-κB-inducing kinase (NIK) expression (median not reached vs 14 months, HR 0.45), both associated with non-canonical NF-κB pathway activation, PFS was significantly longer with IRd vs placebo-Rd. TRAF3 expression was decreased in patients harboring t(4;14) and 1q21 amplification, suggesting increased non-canonical NF-κB pathway activation. CONCLUSIONS Adding ixazomib to Rd provides clinical benefit in MM tumors with increased non-canonical NF-κB pathway activity. This is a potential mechanism for activity in 1q21 amplified high-risk tumors.
Collapse
Affiliation(s)
- Ajeeta B. Dash
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| | - Jacob Zhang
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| | - Lei Shen
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| | - Bin Li
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| | - Deborah Berg
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| | - Jianchang Lin
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| | | | | | - Philippe Moreau
- Department of HematologyUniversity Hospital Hôtel DieuUniversity of NantesNantesFrance
| | | | - Alessandra Di Bacco
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| |
Collapse
|
39
|
Sivakumar P, Thompson JR, Ammar R, Porteous M, McCoubrey C, Cantu E, Ravi K, Zhang Y, Luo Y, Streltsov D, Beers MF, Jarai G, Christie JD. RNA sequencing of transplant-stage idiopathic pulmonary fibrosis lung reveals unique pathway regulation. ERJ Open Res 2019; 5:00117-2019. [PMID: 31423451 PMCID: PMC6689672 DOI: 10.1183/23120541.00117-2019] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/15/2019] [Indexed: 11/05/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF), the scarring of lung parenchyma resulting in the loss of lung function, remains a fatal disease with a significant unmet medical need. Patients with severe IPF often develop acute exacerbations resulting in the rapid deterioration of lung function, requiring transplantation. Understanding the pathophysiological mechanisms contributing to IPF is key to develop novel therapeutic approaches for end-stage disease. We report here RNA-sequencing analyses of lung tissues from a cohort of patients with transplant-stage IPF (n=36), compared with acute lung injury (ALI) (n=11) and nondisease controls (n=19), that reveal a robust gene expression signature unique to end-stage IPF. In addition to extracellular matrix remodelling pathways, we identified pathways associated with T-cell infiltration/activation, tumour development, and cholesterol homeostasis, as well as novel alternatively spliced transcripts that are differentially regulated in the advanced IPF lung versus ALI or nondisease controls. Additionally, we show a subset of genes that are correlated with percent predicted forced vital capacity and could reflect disease severity. Our results establish a robust transcriptomic fingerprint of an advanced IPF lung that is distinct from previously reported microarray signatures of moderate, stable or progressive IPF and identifies hitherto unknown candidate targets and pathways for therapeutic intervention in late-stage IPF as well as biomarkers to characterise disease progression and enable patient stratification.
Collapse
Affiliation(s)
- Pitchumani Sivakumar
- Fibrosis Translational Research and Development, Bristol-Myers Squibb Research and Development, Princeton NJ, USA
| | - John Ryan Thompson
- Translational Bioinformatics, Bristol-Myers Squibb Research and Development, Princeton NJ, USA
| | - Ron Ammar
- Translational Bioinformatics, Bristol-Myers Squibb Research and Development, Princeton NJ, USA
| | - Mary Porteous
- Pulmonary and Critical Care Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Carly McCoubrey
- Pulmonary and Critical Care Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Edward Cantu
- Surgery Dept, University of Pennsylvania, Philadelphia PA, USA
| | - Kandasamy Ravi
- Integrated Genomics, Bristol-Myers Squibb Research and Development, Princeton NJ, USA
| | - Yan Zhang
- Integrated Genomics, Bristol-Myers Squibb Research and Development, Princeton NJ, USA
| | - Yi Luo
- Clinical Biomarkers, Bristol-Myers Squibb Research and Development, Princeton NJ, USA
| | - Denis Streltsov
- Fibrosis Translational Research and Development, Bristol-Myers Squibb Research and Development, Princeton NJ, USA
| | - Michael F Beers
- Pulmonary and Critical Care Medicine, University of Pennsylvania, Philadelphia PA, USA.,PENN Center for Pulmonary Biology, University of Pennsylvania, Philadelphia PA, USA
| | - Gabor Jarai
- Fibrosis Translational Research and Development, Bristol-Myers Squibb Research and Development, Princeton NJ, USA
| | - Jason D Christie
- Pulmonary and Critical Care Medicine, University of Pennsylvania, Philadelphia PA, USA.,PENN Center for Pulmonary Biology, University of Pennsylvania, Philadelphia PA, USA
| |
Collapse
|
40
|
Matsuda-Lennikov M, Biancalana M, Zou J, Ravell JC, Zheng L, Kanellopoulou C, Jiang P, Notarangelo G, Jing H, Masutani E, Oler AJ, Olano LR, Schulz BL, Lenardo MJ. Magnesium transporter 1 (MAGT1) deficiency causes selective defects in N-linked glycosylation and expression of immune-response genes. J Biol Chem 2019; 294:13638-13656. [PMID: 31337704 DOI: 10.1074/jbc.ra119.008903] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/08/2019] [Indexed: 12/22/2022] Open
Abstract
Magnesium transporter 1 (MAGT1) critically mediates magnesium homeostasis in eukaryotes and is highly-conserved across different evolutionary branches. In humans, loss-of-function mutations in the MAGT1 gene cause X-linked magnesium deficiency with Epstein-Barr virus (EBV) infection and neoplasia (XMEN), a disease that has a broad range of clinical and immunological consequences. We have previously shown that EBV susceptibility in XMEN is associated with defective expression of the antiviral natural-killer group 2 member D (NKG2D) protein and abnormal Mg2+ transport. New evidence suggests that MAGT1 is the human homolog of the yeast OST3/OST6 proteins that form an integral part of the N-linked glycosylation complex, although the exact contributions of these perturbations in the glycosylation pathway to disease pathogenesis are still unknown. Using MS-based glycoproteomics, along with CRISPR/Cas9-KO cell lines, natural killer cell-killing assays, and RNA-Seq experiments, we now demonstrate that humans lacking functional MAGT1 have a selective deficiency in both immune and nonimmune glycoproteins, and we identified several critical glycosylation defects in important immune-response proteins and in the expression of genes involved in immunity, particularly CD28. We show that MAGT1 function is partly interchangeable with that of the paralog protein tumor-suppressor candidate 3 (TUSC3) but that each protein has a different tissue distribution in humans. We observed that MAGT1-dependent glycosylation is sensitive to Mg2+ levels and that reduced Mg2+ impairs immune-cell function via the loss of specific glycoproteins. Our findings reveal that defects in protein glycosylation and gene expression underlie immune defects in an inherited disease due to MAGT1 deficiency.
Collapse
Affiliation(s)
- Mami Matsuda-Lennikov
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, NIAID, National Institutes of Health, Bethesda, Maryland 20892.,Clinical Genomics Program, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Matthew Biancalana
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, NIAID, National Institutes of Health, Bethesda, Maryland 20892.,Clinical Genomics Program, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Juan Zou
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, NIAID, National Institutes of Health, Bethesda, Maryland 20892.,Clinical Genomics Program, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Juan C Ravell
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, NIAID, National Institutes of Health, Bethesda, Maryland 20892.,Clinical Genomics Program, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Lixin Zheng
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, NIAID, National Institutes of Health, Bethesda, Maryland 20892.,Clinical Genomics Program, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Chrysi Kanellopoulou
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, NIAID, National Institutes of Health, Bethesda, Maryland 20892.,Clinical Genomics Program, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Ping Jiang
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, NIAID, National Institutes of Health, Bethesda, Maryland 20892.,Clinical Genomics Program, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Giulia Notarangelo
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, NIAID, National Institutes of Health, Bethesda, Maryland 20892.,Clinical Genomics Program, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Huie Jing
- Clinical Genomics Program, NIAID, National Institutes of Health, Bethesda, Maryland 20892.,Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Evan Masutani
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, NIAID, National Institutes of Health, Bethesda, Maryland 20892.,Clinical Genomics Program, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Andrew J Oler
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Lisa Renee Olano
- Laboratory of Neurotoxicology, National Institute of Mental Health, Bethesda, Maryland 20892
| | - Benjamin L Schulz
- University of Queensland, School of Chemistry and Molecular Biology, Brisbane, St. Lucia, Queensland 4072, Australia
| | - Michael J Lenardo
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, NIAID, National Institutes of Health, Bethesda, Maryland 20892 .,Clinical Genomics Program, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
41
|
Connor-Robson N, Booth H, Martin JG, Gao B, Li K, Doig N, Vowles J, Browne C, Klinger L, Juhasz P, Klein C, Cowley SA, Bolam P, Hirst W, Wade-Martins R. An integrated transcriptomics and proteomics analysis reveals functional endocytic dysregulation caused by mutations in LRRK2. Neurobiol Dis 2019; 127:512-526. [PMID: 30954703 PMCID: PMC6597903 DOI: 10.1016/j.nbd.2019.04.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/20/2019] [Accepted: 04/03/2019] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Mutations in LRRK2 are the most common cause of autosomal dominant Parkinson's disease, and the relevance of LRRK2 to the sporadic form of the disease is becoming ever more apparent. It is therefore essential that studies are conducted to improve our understanding of the cellular role of this protein. Here we use multiple models and techniques to identify the pathways through which LRRK2 mutations may lead to the development of Parkinson's disease. METHODS A novel integrated transcriptomics and proteomics approach was used to identify pathways that were significantly altered in iPSC-derived dopaminergic neurons carrying the LRRK2-G2019S mutation. Western blotting, immunostaining and functional assays including FM1-43 analysis of synaptic vesicle endocytosis were performed to confirm these findings in iPSC-derived dopaminergic neuronal cultures carrying either the LRRK2-G2019S or the LRRK2-R1441C mutation, and LRRK2 BAC transgenic rats, and post-mortem human brain tissue from LRRK2-G2019S patients. RESULTS Our integrated -omics analysis revealed highly significant dysregulation of the endocytic pathway in iPSC-derived dopaminergic neurons carrying the LRRK2-G2019S mutation. Western blot analysis confirmed that key endocytic proteins including endophilin I-III, dynamin-1, and various RAB proteins were downregulated in these cultures and in cultures carrying the LRRK2-R1441C mutation, compared with controls. We also found changes in expression of 25 RAB proteins. Changes in endocytic protein expression led to a functional impairment in clathrin-mediated synaptic vesicle endocytosis. Further to this, we found that the endocytic pathway was also perturbed in striatal tissue of aged LRRK2 BAC transgenic rats overexpressing either the LRRK2 wildtype, LRRK2-R1441C or LRRK2-G2019S transgenes. Finally, we found that clathrin heavy chain and endophilin I-III levels are increased in human post-mortem tissue from LRRK2-G2019S patients compared with controls. CONCLUSIONS Our study demonstrates extensive alterations across the endocytic pathway associated with LRRK2 mutations in iPSC-derived dopaminergic neurons and BAC transgenic rats, as well as in post-mortem brain tissue from PD patients carrying a LRRK2 mutation. In particular, we find evidence of disrupted clathrin-mediated endocytosis and suggest that LRRK2-mediated PD pathogenesis may arise through dysregulation of this process.
Collapse
Affiliation(s)
- Natalie Connor-Robson
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| | - Heather Booth
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | | | | | - Natalie Doig
- Medical Research Council Brain Network Dynamics Unit, University of Oxford, Oxford, UK.
| | - Jane Vowles
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK.; Oxford Parkinson's Disease Centre (OPDC), Oxford, UK.
| | - Cathy Browne
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK..
| | - Laura Klinger
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| | | | - Christine Klein
- Institute of Neurogenetics, University of Leubeck, Maria-Goeppert-Str. 1, 23562 Luebeck, Germany..
| | - Sally A Cowley
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK.; Oxford Parkinson's Disease Centre (OPDC), Oxford, UK.
| | - Paul Bolam
- Medical Research Council Brain Network Dynamics Unit, University of Oxford, Oxford, UK; Oxford Parkinson's Disease Centre (OPDC), Oxford, UK.
| | | | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
42
|
Wu C, Huang BE, Chen G, Lovenberg TW, Pocalyko DJ, Yao X. Integrative Analysis of DiseaseLand Omics Database for Disease Signatures and Treatments: A Bipolar Case Study. Front Genet 2019; 10:396. [PMID: 31114610 PMCID: PMC6503737 DOI: 10.3389/fgene.2019.00396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 04/11/2019] [Indexed: 11/13/2022] Open
Abstract
Transcriptomics technologies such as next-generation sequencing and microarray platforms provide exciting opportunities for improving diagnosis and treatment of complex diseases. Transcriptomics studies often share similar hypotheses, but are carried out on different platforms, in different conditions, and with different analysis approaches. These factors, in addition to small sample sizes, can result in a lack of reproducibility. A clear understanding and unified picture of many complex diseases are still elusive, highlighting an urgent need to effectively integrate multiple transcriptomic studies for disease signatures. We have integrated more than 3,000 high-quality transcriptomic datasets in oncology, immunology, neuroscience, cardiovascular and metabolic disease, and from both public and internal sources (DiseaseLand database). We established a systematic data integration and meta-analysis approach, which can be applied in multiple disease areas to create a unified picture of the disease signature and prioritize drug targets, pathways, and compounds. In this bipolar case study, we provided an illustrative example using our approach to combine a total of 30 genome-wide gene expression studies using postmortem human brain samples. First, the studies were integrated by extracting raw FASTQ or CEL files, then undergoing the same procedures for preprocessing, normalization, and statistical inference. Second, both p-value and effect size based meta-analysis algorithms were used to identify a total of 204 differentially expressed (DE) genes (FDR < 0.05) genes in the prefrontal cortex. Among these were BDNF, VGF, WFS1, DUSP6, CRHBP, MAOA, and RELN, which have previously been implicated in bipolar disorder. Finally, pathway enrichment analysis revealed a role for GPCR, MAPK, immune, and Reelin pathways. Compound profiling analysis revealed MAPK and other inhibitors may modulate the DE genes. The ability to robustly combine and synthesize the information from multiple studies enables a more powerful understanding of this complex disease.
Collapse
Affiliation(s)
- Chun Wu
- Computational Sciences, Discovery Sciences, Janssen Research & Development, LLC, Spring House, PA, United States
| | - Bevan E Huang
- Computational Sciences, Discovery Sciences, Janssen Research & Development, LLC, Spring House, PA, United States
| | - Guang Chen
- Neuroscience Therapeutic Area, Janssen Research & Development, LLC, La Jolla, CA, United States
| | - Timothy W Lovenberg
- Neuroscience Therapeutic Area, Janssen Research & Development, LLC, La Jolla, CA, United States
| | - David J Pocalyko
- Computational Sciences, Discovery Sciences, Janssen Research & Development, LLC, Spring House, PA, United States
| | - Xiang Yao
- Computational Sciences, Discovery Sciences, Janssen Research & Development, LLC, La Jolla, CA, United States
| |
Collapse
|
43
|
Zhang H, Liu S, Chang H, Zhan M, Qin QM, Zhang B, Li Z, Liu Y. Mining Magnaporthe oryzae sRNAs With Potential Transboundary Regulation of Rice Genes Associated With Growth and Defense Through Expression Profile Analysis of the Pathogen-Infected Rice. Front Genet 2019; 10:296. [PMID: 30984250 PMCID: PMC6449695 DOI: 10.3389/fgene.2019.00296] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/19/2019] [Indexed: 02/04/2023] Open
Abstract
In recent years, studies have shown that phytopathogenic fungi possess the ability of cross-kingdom regulation of host plants through small RNAs (sRNAs). Magnaporthe oryzae, a causative agent of rice blast, introduces disease by penetrating the rice tissues through appressoria. However, little is known about the transboundary regulation of M. oryzae sRNAs during the interaction of the pathogen with its host rice. Therefore, investigation of the regulation of M. oryzae through sRNAs in the infected rice plants has important theoretical and practical significance for disease control and production improvement. Based on the high-throughput data of M. oryzae sRNAs and the mixed sRNAs during infection, the differential expressions of sRNAs in M. oryzae before and during infection were compared, it was found that expression levels of 366 M. oryzae sRNAs were upregulated significantly during infection. We trained a SVM model which can be used to predict differentially expressed sRNAs, which has reference significance for the prediction of differentially expressed sRNAs of M. oryzae homologous species, and can facilitate the research of M. oryzae in the future. Furthermore, fifty core targets were selected from the predicted target genes on rice for functional enrichment analysis, the analysis reveals that there are nine biological processes and one KEGG pathway associated with rice growth and disease defense. These functions correspond to thirteen rice genes. A total of fourteen M. oryzae sRNAs targeting the rice genes were identified by data analysis, and their authenticity was verified in the database of M. oryzae sRNAs. The 14 M. oryzae sRNAs may participate in the transboundary regulation process and act as sRNA effectors to manipulate the rice blast process.
Collapse
Affiliation(s)
- Hao Zhang
- Key Laboratory of Symbolic Computation and Knowledge Engineering, College of Computer Science and Technology, Ministry of Education, Jilin University, Changchun, China
| | - Sifei Liu
- Key Laboratory of Symbolic Computation and Knowledge Engineering, College of Computer Science and Technology, Ministry of Education, Jilin University, Changchun, China
| | - Haowu Chang
- Key Laboratory of Symbolic Computation and Knowledge Engineering, College of Computer Science and Technology, Ministry of Education, Jilin University, Changchun, China
| | - Mengping Zhan
- Key Laboratory of Symbolic Computation and Knowledge Engineering, College of Computer Science and Technology, Ministry of Education, Jilin University, Changchun, China
| | - Qing-Ming Qin
- College of Plant Sciences, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, China
| | - Borui Zhang
- Columbia Independent School, Columbia, MO, United States
| | - Zhi Li
- School of Computer Science and Technology, Changchun University of Science and Technology, Changchun, China
| | - Yuanning Liu
- Key Laboratory of Symbolic Computation and Knowledge Engineering, College of Computer Science and Technology, Ministry of Education, Jilin University, Changchun, China
| |
Collapse
|
44
|
Lu D, Yamawaki T, Zhou H, Chou WY, Chhoa M, Lamas E, Escobar SS, Arnett HA, Ge H, Juan T, Wang S, Li CM. Limited differential expression of miRNAs and other small RNAs in LPS-stimulated human monocytes. PLoS One 2019; 14:e0214296. [PMID: 30908559 PMCID: PMC6433273 DOI: 10.1371/journal.pone.0214296] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/11/2019] [Indexed: 01/01/2023] Open
Abstract
Monocytes are a distinct subset of myeloid cells with diverse functions in early inflammatory immune modulation. While previous studies have surveyed the role of miRNA regulation on different myeloid cell lines and primary cultures, the time-dependent kinetics of inflammatory stimulation on miRNA expression and the relationship between miRNA-to-target RNA expression have not been comprehensively profiled in monocytes. In this study, we use next-generation sequencing and RT-PCR assays to analyze the non-coding small RNA transcriptome of unstimulated and lipopolysaccharide (LPS)-stimulated monocytes at 6 and 24 hours. We identified a miRNA signature consisting of five mature miRNAs (hsa-mir-146a, hsa-mir-155, hsa-mir-9, hsa-mir-147b, and hsa-mir-193a) upregulated by LPS-stimulated monocytes after 6 hours and found that most miRNAs were also upregulated after 24 hours of stimulation. Only one miRNA gene was down-regulated and no other small RNAs were found dysregulated in monocytes after LPS treatment. In addition, novel tRNA-derived fragments were also discovered in monocytes although none showed significant changes upon LPS stimulation. Interrogation of validated miRNA targets by transcriptomic analysis revealed that absolute expression of most miRNA targets implicating in innate immune response decreased over time in LPS-stimulated monocytes although their expression patterns along the treatment were heterogeneous. Our findings reveal a potential role by which selective miRNA upregulation and stable expression of other small RNAs enable monocytes to develop finely tuned cellular responses during acute inflammation.
Collapse
Affiliation(s)
- Daniel Lu
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Tracy Yamawaki
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Hong Zhou
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Wen-Yu Chou
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Mark Chhoa
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Edwin Lamas
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Sabine S. Escobar
- Inflammation/Oncology TA, Amgen Research, South San Francisco, California, United States of America
| | - Heather A. Arnett
- Inflammation/Oncology TA, Amgen Research, South San Francisco, California, United States of America
| | - Huanying Ge
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Todd Juan
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Songli Wang
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Chi-Ming Li
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
45
|
Huang X, Li L, Ammar R, Zhang Y, Wang Y, Ravi K, Thompson J, Jarai G. Molecular characterization of a precision-cut rat lung slice model for the evaluation of antifibrotic drugs. Am J Physiol Lung Cell Mol Physiol 2019; 316:L348-L357. [DOI: 10.1152/ajplung.00339.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The translation of novel pulmonary fibrosis therapies from preclinical models into the clinic represents a major challenge demonstrated by the high attrition rate of compounds that showed efficacy in preclinical models but demonstrated no significant beneficial effects in clinical trials. A precision-cut lung tissue slice (PCLS) contains all major cell types of the lung and preserves the original cell-cell and cell-matrix contacts. It represents a promising ex vivo model to study pulmonary fibrosis. In this study, using RNA sequencing, we demonstrated that transforming growth factor-β1 (TGFβ1) induced robust fibrotic responses in the rat PCLS model, as it changed the expression of genes functionally related to extracellular matrix remodeling, cell adhesion, epithelial-to-mesenchymal transition, and various immune responses. Nintedanib, pirfenidone, and sorafenib each reversed a subset of genes modulated by TGFβ1, and of those genes we identified 229 whose expression was reversed by all three drugs. These genes define a molecular signature characterizing many aspects of pulmonary fibrosis pathology and its attenuation in the rat PCLS fibrosis model. A panel of 12 genes and three secreted biomarkers, including procollagen I, hyaluronic acid, and WNT1-inducible signaling pathway protein 1 were validated as efficacy end points for the evaluation of antifibrotic activity of experimental compounds. Finally, we showed that blockade of αV-integrins suppressed TGFβ1-induced fibrotic responses in the rat PCLS fibrosis model. Overall, our results suggest that the TGFβ1-induced rat PCLS fibrosis model may represent a valuable system for target validation and to determine the efficacy of experimental compounds.
Collapse
Affiliation(s)
- Xinqiang Huang
- Department of Cardiovascular and Fibrotic Diseases Drug Discovery, Bristol-Myers Squibb, Pennington, New Jersey
| | - Li Li
- Department of Cardiovascular and Fibrotic Diseases Drug Discovery, Bristol-Myers Squibb, Pennington, New Jersey
| | - Ron Ammar
- Department of Cardiovascular and Fibrotic Diseases Drug Discovery, Bristol-Myers Squibb, Pennington, New Jersey
| | - Yan Zhang
- Department of Cardiovascular and Fibrotic Diseases Drug Discovery, Bristol-Myers Squibb, Pennington, New Jersey
| | - Yihe Wang
- Department of Cardiovascular and Fibrotic Diseases Drug Discovery, Bristol-Myers Squibb, Pennington, New Jersey
| | - Kandasamy Ravi
- Department of Cardiovascular and Fibrotic Diseases Drug Discovery, Bristol-Myers Squibb, Pennington, New Jersey
| | - John Thompson
- Department of Cardiovascular and Fibrotic Diseases Drug Discovery, Bristol-Myers Squibb, Pennington, New Jersey
| | - Gabor Jarai
- Department of Cardiovascular and Fibrotic Diseases Drug Discovery, Bristol-Myers Squibb, Pennington, New Jersey
| |
Collapse
|
46
|
Rao MS, Van Vleet TR, Ciurlionis R, Buck WR, Mittelstadt SW, Blomme EAG, Liguori MJ. Comparison of RNA-Seq and Microarray Gene Expression Platforms for the Toxicogenomic Evaluation of Liver From Short-Term Rat Toxicity Studies. Front Genet 2019; 9:636. [PMID: 30723492 PMCID: PMC6349826 DOI: 10.3389/fgene.2018.00636] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/27/2018] [Indexed: 12/12/2022] Open
Abstract
Gene expression profiling is a useful tool to predict and interrogate mechanisms of toxicity. RNA-Seq technology has emerged as an attractive alternative to traditional microarray platforms for conducting transcriptional profiling. The objective of this work was to compare both transcriptomic platforms to determine whether RNA-Seq offered significant advantages over microarrays for toxicogenomic studies. RNA samples from the livers of rats treated for 5 days with five tool hepatotoxicants (α-naphthylisothiocyanate/ANIT, carbon tetrachloride/CCl4, methylenedianiline/MDA, acetaminophen/APAP, and diclofenac/DCLF) were analyzed with both gene expression platforms (RNA-Seq and microarray). Data were compared to determine any potential added scientific (i.e., better biological or toxicological insight) value offered by RNA-Seq compared to microarrays. RNA-Seq identified more differentially expressed protein-coding genes and provided a wider quantitative range of expression level changes when compared to microarrays. Both platforms identified a larger number of differentially expressed genes (DEGs) in livers of rats treated with ANIT, MDA, and CCl4 compared to APAP and DCLF, in agreement with the severity of histopathological findings. Approximately 78% of DEGs identified with microarrays overlapped with RNA-Seq data, with a Spearman’s correlation of 0.7 to 0.83. Consistent with the mechanisms of toxicity of ANIT, APAP, MDA and CCl4, both platforms identified dysregulation of liver relevant pathways such as Nrf2, cholesterol biosynthesis, eiF2, hepatic cholestasis, glutathione and LPS/IL-1 mediated RXR inhibition. RNA-Seq data showed additional DEGs that not only significantly enriched these pathways, but also suggested modulation of additional liver relevant pathways. In addition, RNA-Seq enabled the identification of non-coding DEGs that offer a potential for improved mechanistic clarity. Overall, these results indicate that RNA-Seq is an acceptable alternative platform to microarrays for rat toxicogenomic studies with several advantages. Because of its wider dynamic range as well as its ability to identify a larger number of DEGs, RNA-Seq may generate more insight into mechanisms of toxicity. However, more extensive reference data will be necessary to fully leverage these additional RNA-Seq data, especially for non-coding sequences.
Collapse
Affiliation(s)
- Mohan S Rao
- Investigative Toxicology and Pathology, Global Preclinical Safety, AbbVie, North Chicago, IL, United States
| | - Terry R Van Vleet
- Investigative Toxicology and Pathology, Global Preclinical Safety, AbbVie, North Chicago, IL, United States
| | - Rita Ciurlionis
- Investigative Toxicology and Pathology, Global Preclinical Safety, AbbVie, North Chicago, IL, United States
| | - Wayne R Buck
- Investigative Toxicology and Pathology, Global Preclinical Safety, AbbVie, North Chicago, IL, United States
| | - Scott W Mittelstadt
- Investigative Toxicology and Pathology, Global Preclinical Safety, AbbVie, North Chicago, IL, United States
| | - Eric A G Blomme
- Investigative Toxicology and Pathology, Global Preclinical Safety, AbbVie, North Chicago, IL, United States
| | - Michael J Liguori
- Investigative Toxicology and Pathology, Global Preclinical Safety, AbbVie, North Chicago, IL, United States
| |
Collapse
|
47
|
Huang X, Cai H, Ammar R, Zhang Y, Wang Y, Ravi K, Thompson J, Jarai G. Molecular characterization of a precision-cut rat liver slice model for the evaluation of antifibrotic compounds. Am J Physiol Gastrointest Liver Physiol 2019; 316:G15-G24. [PMID: 30406699 PMCID: PMC6383382 DOI: 10.1152/ajpgi.00281.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Precision-cut liver tissue slice (PCLS) contains all major cell types of the liver parenchyma and preserves the original cell-cell and cell-matrix contacts. It represents a promising ex vivo model to study liver fibrosis and test the antifibrotic effect of experimental compounds in a physiological environment. In this study using RNA sequencing, we demonstrated that various pathways functionally related to fibrotic mechanisms were dysregulated in PCLSs derived from rats subjected to bile duct ligation. The activin receptor-like kinase-5 (Alk5) inhibitor SB525334, nintedanib, and sorafenib each reversed a subset of genes dysregulated in fibrotic PCLSs, and of those genes we identified 608 genes whose expression was reversed by all three compounds. These genes define a molecular signature characterizing many aspects of liver fibrosis pathology and its attenuation in the model. A panel of 12 genes and 4 secreted biomarkers including procollagen I, hyaluronic acid (HA), insulin-like growth factor binding protein 5 (IGFBP5), and WNT1-inducible signaling pathway protein 1 (WISP1) were further validated as efficacy end points for the evaluation of antifibrotic activity of experimental compounds. Finally, we showed that blockade of αV-integrins with a small molecule inhibitor attenuated the fibrotic phenotype in the model. Overall, our results suggest that the rat fibrotic PCLS model may represent a valuable system for target validation and determining the efficacy of experimental compounds. NEW & NOTEWORTHY We investigated the antifibrotic activity of three compounds, the activin receptor-like kinase-5 (Alk5) inhibitor SB525334, nintedanib, and sorafenib, in a rat fibrotic precision-cut liver tissue slice model using RNA sequencing analysis. A panel of 12 genes and 4 secreted biomarkers including procollagen I, hyaluronic acid (HA), insulin-like growth factor binding protein 5 (IGFBP5), and WNT1-inducible signaling pathway protein 1 (WISP1) were then established as efficacy end points to validate the antifibrotic activity of the αV-integrin inhibitor CWHM12. This study demonstrated the value of the rat fibrotic PCLS model for the evaluation of antifibrotic drugs.
Collapse
Affiliation(s)
| | - Hong Cai
- Bristol-Myers Squibb, Pennington, New Jersey
| | - Ron Ammar
- Bristol-Myers Squibb, Pennington, New Jersey
| | - Yan Zhang
- Bristol-Myers Squibb, Pennington, New Jersey
| | - Yihe Wang
- Bristol-Myers Squibb, Pennington, New Jersey
| | | | | | - Gabor Jarai
- Bristol-Myers Squibb, Pennington, New Jersey
| |
Collapse
|
48
|
Pitt JJ, Riester M, Zheng Y, Yoshimatsu TF, Sanni A, Oluwasola O, Veloso A, Labrot E, Wang S, Odetunde A, Ademola A, Okedere B, Mahan S, Leary R, Macomber M, Ajani M, Johnson RS, Fitzgerald D, Grundstad AJ, Tuteja JH, Khramtsova G, Zhang J, Sveen E, Hwang B, Clayton W, Nkwodimmah C, Famooto B, Obasi E, Aderoju V, Oludara M, Omodele F, Akinyele O, Adeoye A, Ogundiran T, Babalola C, MacIsaac K, Popoola A, Morrissey MP, Chen LS, Wang J, Olopade CO, Falusi AG, Winckler W, Haase K, Van Loo P, Obafunwa J, Papoutsakis D, Ojengbede O, Weber B, Ibrahim N, White KP, Huo D, Olopade OI, Barretina J. Characterization of Nigerian breast cancer reveals prevalent homologous recombination deficiency and aggressive molecular features. Nat Commun 2018; 9:4181. [PMID: 30327465 PMCID: PMC6191428 DOI: 10.1038/s41467-018-06616-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 09/13/2018] [Indexed: 02/08/2023] Open
Abstract
Racial/ethnic disparities in breast cancer mortality continue to widen but genomic studies rarely interrogate breast cancer in diverse populations. Through genome, exome, and RNA sequencing, we examined the molecular features of breast cancers using 194 patients from Nigeria and 1037 patients from The Cancer Genome Atlas (TCGA). Relative to Black and White cohorts in TCGA, Nigerian HR + /HER2 - tumors are characterized by increased homologous recombination deficiency signature, pervasive TP53 mutations, and greater structural variation-indicating aggressive biology. GATA3 mutations are also more frequent in Nigerians regardless of subtype. Higher proportions of APOBEC-mediated substitutions strongly associate with PIK3CA and CDH1 mutations, which are underrepresented in Nigerians and Blacks. PLK2, KDM6A, and B2M are also identified as previously unreported significantly mutated genes in breast cancer. This dataset provides novel insights into potential molecular mechanisms underlying outcome disparities and lay a foundation for deployment of precision therapeutics in underserved populations.
Collapse
Affiliation(s)
- Jason J Pitt
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore
| | - Markus Riester
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Yonglan Zheng
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Toshio F Yoshimatsu
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Ayodele Sanni
- Department of Pathology and Forensic Medicine, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | | | - Artur Veloso
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Emma Labrot
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Shengfeng Wang
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, 100191, China
| | - Abayomi Odetunde
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Adeyinka Ademola
- Department of Surgery, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Babajide Okedere
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Scott Mahan
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Rebecca Leary
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Maura Macomber
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Mustapha Ajani
- Department of Pathology, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Ryan S Johnson
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Dominic Fitzgerald
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - A Jason Grundstad
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Jigyasa H Tuteja
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Galina Khramtsova
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Jing Zhang
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Elisabeth Sveen
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Bryce Hwang
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Wendy Clayton
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | | - Bisola Famooto
- Department of Surgery, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Esther Obasi
- Department of Pathology and Forensic Medicine, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Victor Aderoju
- Department of Surgery, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Mobolaji Oludara
- Department of Surgery, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Folusho Omodele
- Department of Surgery, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Odunayo Akinyele
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Adewunmi Adeoye
- Department of Pathology, University of Ibadan, Ibadan, Oyo, Nigeria
| | | | - Chinedum Babalola
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
- Department of Pharmaceutical Chemistry, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Kenzie MacIsaac
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Abiodun Popoola
- Oncology Unit, Department of Radiology, Lagos State University, Ikeja, Lagos, Nigeria
| | | | - Lin S Chen
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Jiebiao Wang
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Christopher O Olopade
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Adeyinka G Falusi
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Wendy Winckler
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Kerstin Haase
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Peter Van Loo
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Department of Human Genetics, University of Leuven, Oude Markt 13, Leuven, 3000, Belgium
| | - John Obafunwa
- Department of Pathology and Forensic Medicine, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | | | - Oladosu Ojengbede
- Centre for Population and Reproductive Health, College of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Barbara Weber
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Nasiru Ibrahim
- Department of Surgery, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Kevin P White
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA.
- Tempus Labs Inc., Chicago, IL, USA.
| | - Dezheng Huo
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA.
| | - Olufunmilayo I Olopade
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA.
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| | - Jordi Barretina
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA.
- Girona Biomedical Research Institute (IDIBGI), Girona, 17007, Spain.
| |
Collapse
|
49
|
Williams AL, Khadka V, Tang M, Avelar A, Schunke KJ, Menor M, Shohet RV. HIF1 mediates a switch in pyruvate kinase isoforms after myocardial infarction. Physiol Genomics 2018; 50:479-494. [PMID: 29652636 PMCID: PMC6087881 DOI: 10.1152/physiolgenomics.00130.2017] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 12/20/2022] Open
Abstract
Alternative splicing of RNA is an underexplored area of transcriptional response. We expect that early changes in alternatively spliced genes may be important for responses to cardiac injury. Hypoxia inducible factor 1 (HIF1) is a key transcription factor that rapidly responds to loss of oxygen through alteration of metabolism and angiogenesis. The goal of this study was to investigate the transcriptional response after myocardial infarction (MI) and to identify novel, hypoxia-driven changes, including alternative splicing. After ligation of the left anterior descending artery in mice, we observed an abrupt loss of cardiac contractility and upregulation of hypoxic signaling. We then performed RNA sequencing on ischemic heart tissue 1 and 3 days after infarct to assess early transcriptional changes and identified 89 transcripts with altered splicing. Of particular interest was the switch in Pkm isoform expression (pyruvate kinase, muscle). The usually predominant Pkm1 isoform was less abundant in ischemic hearts, while Pkm2 and associated splicing factors (hnRNPA1, hnRNPA2B1, Ptbp1) rapidly increased. Despite increased Pkm2 expression, total pyruvate kinase activity remained reduced in ischemic myocardial tissue. We also demonstrated HIF1 binding to PKM by chromatin immunoprecipitation, indicating a direct role for HIF1 in mediating this isoform switch. Our study provides a new, detailed characterization of the early transcriptome after MI. From this analysis, we identified an HIF1-mediated alternative splicing event in the PKM gene. Pkm1 and Pkm2 play distinct roles in glycolytic metabolism and the upregulation of Pkm2 is likely to have important consequences for ATP synthesis in infarcted cardiac muscle.
Collapse
Affiliation(s)
- Allison Lesher Williams
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii , Honolulu, Hawaii
| | - Vedbar Khadka
- Bioinformatics Core, John A. Burns School of Medicine, University of Hawaii , Honolulu, Hawaii
| | - Mingxin Tang
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii , Honolulu, Hawaii
| | - Abigail Avelar
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii , Honolulu, Hawaii
| | - Kathryn J Schunke
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii , Honolulu, Hawaii
| | - Mark Menor
- Bioinformatics Core, John A. Burns School of Medicine, University of Hawaii , Honolulu, Hawaii
| | - Ralph V Shohet
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii , Honolulu, Hawaii
| |
Collapse
|
50
|
He Y, Yao X, Taylor N, Bai Y, Lovenberg T, Bhattacharya A. RNA sequencing analysis reveals quiescent microglia isolation methods from postnatal mouse brains and limitations of BV2 cells. J Neuroinflammation 2018; 15:153. [PMID: 29788964 PMCID: PMC5964710 DOI: 10.1186/s12974-018-1195-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 05/10/2018] [Indexed: 12/22/2022] Open
Abstract
Background Microglia play key roles in neuron–glia interaction, neuroinflammation, neural repair, and neurotoxicity. Currently, various microglial in vitro models including primary microglia derived from distinct isolation methods and immortalized microglial cell lines are extensively used. However, the diversity of these existing models raises difficulty in parallel comparison across studies since microglia are sensitive to environmental changes, and thus, different models are likely to show widely varied responses to the same stimuli. To better understand the involvement of microglia in pathophysiological situations, it is critical to establish a reliable microglial model system. Methods With postnatal mouse brains, we isolated microglia using three general methods including shaking, mild trypsinization, and CD11b magnetic-associated cell sorting (MACS) and applied RNA sequencing to compare transcriptomes of the isolated cells. Additionally, we generated a genome-wide dataset by RNA sequencing of immortalized BV2 microglial cell line to compare with primary microglia. Furthermore, based on the outcomes of transcriptional analysis, we compared cellular functions between primary microglia and BV2 cells including immune responses to LPS by quantitative RT-PCR and Luminex Multiplex Assay, TGFβ signaling probed by Western blot, and direct migration by chemotaxis assay. Results We found that although the yield and purity of microglia were comparable among the three isolation methods, mild trypsinization drove microglia in a relatively active state, evidenced by high amount of amoeboid microglia, enhanced expression of microglial activation genes, and suppression of microglial quiescent genes. In contrast, CD11b MACS was the most reliable and consistent method, and microglia isolated by this method maintained a relatively resting state. Transcriptional and functional analyses revealed that as compared to primary microglia, BV2 cells remain most of the immune functions such as responses to LPS but showed limited TGFβ signaling and chemotaxis upon chemoattractant C5a. Conclusions Collectively, we determined the optimal isolation methods for quiescent microglia and characterized the limitations of BV2 cells as an alternative of primary microglia. Considering transcriptional and functional differences, caution should be taken when extrapolating data from various microglial models. In addition, our RNA sequencing database serves as a valuable resource to provide novel insights for appropriate application of microglia as in vitro models. Electronic supplementary material The online version of this article (10.1186/s12974-018-1195-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yingbo He
- Janssen Research & Development, LLC., Neuroscience Drug Discovery, 3210 Merryfield Row, San Diego, CA, 92121, USA.
| | - Xiang Yao
- Janssen Research & Development, LLC., Discovery Sciences, San Diego, CA, USA
| | - Natalie Taylor
- Janssen Research & Development, LLC., Neuroscience Drug Discovery, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Yuchen Bai
- Janssen Research & Development, LLC., Discovery Sciences, Spring House, PA, USA
| | - Timothy Lovenberg
- Janssen Research & Development, LLC., Neuroscience Drug Discovery, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Anindya Bhattacharya
- Janssen Research & Development, LLC., Neuroscience Drug Discovery, 3210 Merryfield Row, San Diego, CA, 92121, USA
| |
Collapse
|