88 results on '"Rothlin CV"'
Search Results
2. Cholesterol mobilization regulates dendritic cell maturation and the immunogenic response to cancer.
- Author
-
Belabed M, Park MD, Blouin CM, Balan S, Moon CY, Freed G, Quijada-Álamo M, Peros A, Mattiuz R, Reid AM, Yatim N, Boumelha J, Azimi CS, LaMarche NM, Troncoso L, Amabile A, Le Berichel J, Chen ST, Wilk CM, Brown BD, Radford KJ, Ghosh S, Rothlin CV, Yvan-Charvet L, Marron TU, Puleston DJ, Wagenblast E, Bhardwaj N, Lamaze C, and Merad M
- Subjects
- Animals, Mice, Humans, Neoplasms immunology, Proto-Oncogene Proteins metabolism, Mice, Inbred C57BL, Mice, Knockout, Signal Transduction immunology, Dendritic Cells immunology, Dendritic Cells metabolism, Cholesterol metabolism, Cell Differentiation immunology, Axl Receptor Tyrosine Kinase, Receptor Protein-Tyrosine Kinases metabolism, Receptor Protein-Tyrosine Kinases immunology
- Abstract
Maturation of conventional dendritic cells (cDCs) is crucial for maintaining tolerogenic safeguards against auto-immunity and for promoting immunogenic responses to pathogens and cancer. The subcellular mechanism for cDC maturation remains poorly defined. We show that cDCs mature by leveraging an internal reservoir of cholesterol (harnessed from extracellular cell debris and generated by de novo synthesis) to assemble lipid nanodomains on cell surfaces of maturing cDCs, enhance expression of maturation markers and stabilize immune receptor signaling. This process is dependent on cholesterol transport through Niemann-Pick disease type C1 (NPC1) and mediates homeostatic and Toll-like receptor (TLR)-induced maturation. Importantly, we identified the receptor tyrosine kinase AXL as a regulator of the NPC1-dependent construction of lipid nanodomains. Deleting AXL from cDCs enhances their maturation, thus improving anti-tumor immunity. Altogether, our study presents new insights into cholesterol mobilization as a fundamental basis for cDC maturation and highlights AXL as a therapeutic target for modulating cDCs., Competing Interests: Competing interests: M.M. serves on the scientific advisory board of and holds stock from Compugen, Dynavax, Innate Pharma, Morphic Therapeutics, Asher Bio, Dren Bio, Nirogy, Genenta, OncoResponse and Owkin. M.M. serves on the ad hoc scientific advisory board of DBV and Genentech and on the foundation advisory board of Breakthrough Cancer. M.M. receives funding for contracted research from Genentech, Regeneron and Boehringer Ingelheim. M.M. is listed as an inventor on a patent application (16/092576) submitted by the Icahn School of Medicine at Mount Sinai that covers the use of multiplex immunohistochemistry to characterize tumors and treatment responses. The technology is filed through the Icahn School of Medicine at Mount Sinai and is currently unlicensed. This technology was used to evaluate tissue in this study, and the results could impact the value of this technology. T.U.M. has served on advisory and/or data safety monitoring boards for Rockefeller University, Regeneron Pharmaceuticals, AbbVie, Bristol Meyers Squibb, Boehringer Ingelheim, Atara, AstraZeneca, Genentech, Celldex, Chimeric, Glenmark, Simcere, Surface, G1 Therapeutics, NGM Bio, DBV Technologies, Arcus and Astellas and has research grants from Regeneron, Bristol Myers Squibb, Merck and Boehringer Ingelheim. The remaining authors declare no competing interests., (© 2025. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2025
- Full Text
- View/download PDF
3. Feeding the wrath with myelin.
- Author
-
Ghosh S and Rothlin CV
- Subjects
- Humans, Animals, Macrophages immunology, Macrophages metabolism, Brain Neoplasms immunology, Brain Neoplasms pathology, Brain Neoplasms metabolism, Myelin Sheath metabolism, Myelin Sheath immunology, Glioblastoma immunology, Glioblastoma metabolism, Glioblastoma pathology
- Abstract
Kloosterman and colleagues studied molecular and cellular changes during radiation therapy and disease recurrence across molecular subtypes of glioblastoma. They uncovered a distinct immune-cancer cell metabolic crosstalk during proneural/oligodendrocyte progenitor cell-like to mesenchymal-like transition, wherein macrophages feed on cholesterol-rich myelin debris to provide lipids to mesenchymal tumor cells, thereby fueling glioblastoma growth., Competing Interests: Declaration of interests C.V.R. is a member of the Trends in Immunology Advisory Board., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. Stem cells tightly regulate dead cell clearance to maintain tissue fitness.
- Author
-
Stewart KS, Abdusselamoglu MD, Tierney MT, Gola A, Hur YH, Gonzales KAU, Yuan S, Bonny AR, Yang Y, Infarinato NR, Cowley CJ, Levorse JM, Pasolli HA, Ghosh S, Rothlin CV, and Fuchs E
- Subjects
- Animals, Female, Male, Mice, Ligands, Phagocytes cytology, Phagocytes metabolism, Retinoids metabolism, Lipid Metabolism, Retinoic Acid Receptor gamma metabolism, Retinoid X Receptor alpha metabolism, Apoptosis, Hair Follicle cytology, Hair Follicle metabolism, Hair Follicle pathology, Homeostasis, Phagocytosis, Regeneration, Stem Cells cytology, Stem Cells metabolism
- Abstract
Billions of cells are eliminated daily from our bodies
1-4 . Although macrophages and dendritic cells are dedicated to migrating and engulfing dying cells and debris, many epithelial and mesenchymal tissue cells can digest nearby apoptotic corpses1-4 . How these non-motile, non-professional phagocytes sense and eliminate dying cells while maintaining their normal tissue functions is unclear. Here we explore the mechanisms that underlie their multifunctionality by exploiting the cyclical bouts of tissue regeneration and degeneration during hair cycling. We show that hair follicle stem cells transiently unleash phagocytosis at the correct time and place through local molecular triggers that depend on both lipids released by neighbouring apoptotic corpses and retinoids released by healthy counterparts. We trace the heart of this dual ligand requirement to RARγ-RXRα, whose activation enables tight regulation of apoptotic cell clearance genes and provides an effective, tunable mechanism to offset phagocytic duties against the primary stem cell function of preserving tissue integrity during homeostasis. Finally, we provide functional evidence that hair follicle stem cell-mediated phagocytosis is not simply redundant with professional phagocytes but rather has clear benefits to tissue fitness. Our findings have broad implications for other non-motile tissue stem or progenitor cells that encounter cell death in an immune-privileged niche., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
5. The amalgam of naive CD4 + T cell transcriptional states is reconfigured by helminth infection to dampen the amplitude of the immune response.
- Author
-
Even Z, Meli AP, Tyagi A, Vidyarthi A, Briggs N, de Kouchkovsky DA, Kong Y, Wang Y, Waizman DA, Rice TA, De Kumar B, Wang X, Palm NW, Craft J, Basu MK, Ghosh S, and Rothlin CV
- Subjects
- Animals, Mice, Strongylida Infections immunology, Strongylida Infections parasitology, Specific Pathogen-Free Organisms, Transcription, Genetic, Receptors, Antigen, T-Cell immunology, Receptors, Antigen, T-Cell metabolism, Helminthiasis immunology, Interferon Type I metabolism, Interferon Type I immunology, Mice, Inbred C57BL, Lymphocyte Activation immunology, CD4-Positive T-Lymphocytes immunology, Nippostrongylus immunology
- Abstract
Naive CD4
+ T cells in specific pathogen-free (SPF) mice are characterized by transcriptional heterogeneity and subpopulations distinguished by the expression of quiescence, the extracellular matrix (ECM) and cytoskeleton, type I interferon (IFN-I) response, memory-like, and T cell receptor (TCR) activation genes. We demonstrate that this constitutive heterogeneity, including the presence of the IFN-I response cluster, is commensal independent insofar as being identical in germ-free and SPF mice. By contrast, Nippostrongylus brasiliensis infection altered this constitutive heterogeneity. Naive T cell-intrinsic transcriptional changes acquired during helminth infection correlated with and accounted for decreased immunization response to an unrelated antigen. These compositional and functional changes were dependent variables of helminth infection, as they disappeared at the established time point of its clearance in mice. Collectively, our results indicate that the naive T cell pool is subject to dynamic transcriptional changes in response to certain environmental cues, which in turn permutes the magnitude of the immune response., Competing Interests: Declaration of interests Z.E. is currently a medical student at the University of North Dakota; A.P.M. is a postdoctoral fellow at McGill University; D.A.d.K. is currently a resident at the University of California, San Diego; and Y.W. is currently a postdoctoral fellow at St. Jude Children’s Research Hospital., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
6. In the Eyes of the Beholder-New Mertk Knockout Mouse and Re-Evaluation of Phagocytosis versus Anti-Inflammatory Functions of MERTK.
- Author
-
Ghosh S, Finnemann SC, Vollrath D, and Rothlin CV
- Subjects
- Animals, Mice, Retinal Degeneration genetics, Retinal Degeneration metabolism, Retinal Degeneration pathology, Disease Models, Animal, Receptor Protein-Tyrosine Kinases metabolism, Receptor Protein-Tyrosine Kinases genetics, Humans, Inflammation genetics, Inflammation metabolism, c-Mer Tyrosine Kinase metabolism, c-Mer Tyrosine Kinase genetics, Phagocytosis, Mice, Knockout
- Abstract
Greg Lemke's laboratory was one of the pioneers of research into the TAM family of receptor tyrosine kinases (RTKs). Not only was Tyro3 cloned in his laboratory, but his group also extensively studied mice knocked out for individual or various combinations of the TAM RTKs Tyro3 , Axl , and Mertk . Here we primarily focus on one of the paralogs-MERTK. We provide a historical perspective on rodent models of loss of Mertk function and their association with retinal degeneration and blindness. We describe later studies employing mouse genetics and the generation of newer knockout models that point out incongruencies with the inference that loss of MERTK-dependent phagocytosis is sufficient for severe, early-onset photoreceptor degeneration in mice. This discussion is meant to raise awareness with regards to the limitations of the original Mertk knockout mouse model generated using 129 derived embryonic stem cells and carrying 129 derived alleles and the role of these alleles in modifying Mertk knockout phenotypes or even displaying Mertk -independent phenotypes. We also suggest molecular approaches that can further Greg Lemke's scintillating legacy of dissecting the molecular functions of MERTK-a protein that has been described to function in phagocytosis as well as in the negative regulation of inflammation.
- Published
- 2024
- Full Text
- View/download PDF
7. TREM2 function in glioblastoma immune microenvironment: Can we distinguish reality from illusion?
- Author
-
Ghosh S and Rothlin CV
- Subjects
- Humans, Glioblastoma immunology, Glioblastoma metabolism, Glioblastoma pathology, Tumor Microenvironment immunology, Brain Neoplasms immunology, Brain Neoplasms metabolism, Brain Neoplasms pathology, Receptors, Immunologic metabolism, Receptors, Immunologic immunology, Membrane Glycoproteins metabolism, Membrane Glycoproteins immunology
- Published
- 2024
- Full Text
- View/download PDF
8. Apoptotic cell identity induces distinct functional responses to IL-4 in efferocytic macrophages.
- Author
-
Liebold I, Al Jawazneh A, Casar C, Lanzloth C, Leyk S, Hamley M, Wong MN, Kylies D, Gräfe SK, Edenhofer I, Aranda-Pardos I, Kriwet M, Haas H, Krause J, Hadjilaou A, Schromm AB, Richardt U, Eggert P, Tappe D, Weidemann SA, Ghosh S, Krebs CF, A-Gonzalez N, Worthmann A, Lohse AW, Huber S, Rothlin CV, Puelles VG, Jacobs T, Gagliani N, and Bosurgi L
- Subjects
- Animals, Mice, Hepatocytes immunology, Mice, Knockout, Neutrophils immunology, Disease Models, Animal, Apoptosis immunology, Interleukin-4 genetics, Interleukin-4 metabolism, Macrophages immunology, Phagocytosis immunology, Schistosomiasis mansoni genetics, Schistosomiasis mansoni immunology
- Abstract
Macrophages are functionally heterogeneous cells essential for apoptotic cell clearance. Apoptotic cells are defined by homogeneous characteristics, ignoring their original cell lineage identity. We found that in an interleukin-4 (IL-4)-enriched environment, the sensing of apoptotic neutrophils by macrophages triggered their tissue remodeling signature. Engulfment of apoptotic hepatocytes promoted a tolerogenic phenotype, whereas phagocytosis of T cells had little effect on IL-4-induced gene expression. In a mouse model of parasite-induced pathology, the transfer of macrophages conditioned with IL-4 and apoptotic neutrophils promoted parasitic egg clearance. Knockout of phagocytic receptors required for the uptake of apoptotic neutrophils and partially T cells, but not hepatocytes, exacerbated helminth infection. These findings suggest that the identity of apoptotic cells may contribute to the development of distinct IL-4-driven immune programs in macrophages.
- Published
- 2024
- Full Text
- View/download PDF
9. Nmes1 is a novel regulator of mucosal response influencing intestinal healing potential.
- Author
-
Hamley M, Leyk S, Casar C, Liebold I, Jawazneh AA, Lanzloth C, Böttcher M, Haas H, Richardt U, Rothlin CV, Jacobs T, Huber S, Adlung L, Pelczar P, Henao-Mejia J, and Bosurgi L
- Subjects
- Animals, Mice, Cytokines, Intestines, Wound Healing, Colitis drug therapy, Intestinal Mucosa
- Abstract
The initiation of tissue remodeling following damage is a critical step in preventing the development of immune-mediated diseases. Several factors contribute to mucosal healing, leading to innovative therapeutic approaches for managing intestinal disorders. However, uncovering alternative targets and gaining mechanistic insights are imperative to enhance therapy efficacy and broaden its applicability across different intestinal diseases. Here we demonstrate that Nmes1, encoding for Normal Mucosa of Esophagus-Specific gene 1, also known as Aa467197, is a novel regulator of mucosal healing. Nmes1 influences the macrophage response to the tissue remodeling cytokine IL-4 in vitro. In addition, using two murine models of intestinal damage, each characterized by a type 2-dominated environment with contrasting functions, the ablation of Nmes1 results in decreased intestinal regeneration during the recovery phase of colitis, while enhancing parasitic egg clearance and reducing fibrosis during the advanced stages of Schistosoma mansoni infection. These outcomes are associated with alterations in CX3CR1
+ macrophages, cells known for their wound-healing potential in the inflamed colon, hence promising candidates for cell therapies. All in all, our data indicate Nmes1 as a novel contributor to mucosal healing, setting the basis for further investigation into its potential as a new target for the treatment of colon-associated inflammation., (© 2023 The Authors. European Journal of Immunology published by Wiley-VCH GmbH.)- Published
- 2024
- Full Text
- View/download PDF
10. Age-dependent differences in efferocytosis determine the outcome of opsonophagocytic protection from invasive pathogens.
- Author
-
Bee GCW, Lokken-Toyli KL, Yeung ST, Rodriguez L, Zangari T, Anderson EE, Ghosh S, Rothlin CV, Brodin P, Khanna KM, and Weiser JN
- Subjects
- Mice, Animals, Humans, Macrophages metabolism, Streptococcus pneumoniae, c-Mer Tyrosine Kinase, Phagocytosis, Neutrophils
- Abstract
In early life, susceptibility to invasive infection skews toward a small subset of microbes, whereas other pathogens associated with diseases later in life, including Streptococcus pneumoniae (Spn), are uncommon among neonates. To delineate mechanisms behind age-dependent susceptibility, we compared age-specific mouse models of invasive Spn infection. We show enhanced CD11b-dependent opsonophagocytosis by neonatal neutrophils improved protection against Spn during early life. The augmented function of neonatal neutrophils was mediated by higher CD11b surface expression at the population level due to dampened efferocytosis, which also resulted in more CD11b
hi "aged" neutrophils in peripheral blood. Dampened efferocytosis during early life could be attributed to the lack of CD169+ macrophages in neonates and reduced systemic expressions of multiple efferocytic mediators, including MerTK. On experimentally impairing efferocytosis later in life, CD11bhi neutrophils increased and protection against Spn improved. Our findings reveal how age-dependent differences in efferocytosis determine infection outcome through the modulation of CD11b-driven opsonophagocytosis and immunity., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
11. Stem cells tightly regulate dead cell clearance to maintain tissue fitness.
- Author
-
Stewart KS, Gonzales KA, Yuan S, Tierney MT, Bonny AR, Yang Y, Infarinato NR, Cowley CJ, Levorse JM, Pasolli HA, Ghosh S, Rothlin CV, and Fuchs E
- Abstract
Macrophages and dendritic cells have long been appreciated for their ability to migrate to and engulf dying cells and debris, including some of the billions of cells that are naturally eliminated from our body daily. However, a substantial number of these dying cells are cleared by 'non-professional phagocytes', local epithelial cells that are critical to organismal fitness. How non-professional phagocytes sense and digest nearby apoptotic corpses while still performing their normal tissue functions is unclear. Here, we explore the molecular mechanisms underlying their multifunctionality. Exploiting the cyclical bouts of tissue regeneration and degeneration during the hair cycle, we show that stem cells can transiently become non-professional phagocytes when confronted with dying cells. Adoption of this phagocytic state requires both local lipids produced by apoptotic corpses to activate RXRα, and tissue-specific retinoids for RARγ activation. This dual factor dependency enables tight regulation of the genes requisite to activate phagocytic apoptotic clearance. The tunable phagocytic program we describe here offers an effective mechanism to offset phagocytic duties against the primary stem cell function of replenishing differentiated cells to preserve tissue integrity during homeostasis. Our findings have broad implications for other non-motile stem or progenitor cells which experience cell death in an immune-privileged niche., Competing Interests: Competing Interest E.F. has served on the scientific advisory boards of L’Oreal and Arsenal Biosciences. C.V.R is a senior editor for eLife. S.G. has received grant support from Mirati Therapeutics. The other authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
12. Apoptotic cell death in disease-Current understanding of the NCCD 2023.
- Author
-
Vitale I, Pietrocola F, Guilbaud E, Aaronson SA, Abrams JM, Adam D, Agostini M, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, Aqeilan RI, Arama E, Baehrecke EH, Balachandran S, Bano D, Barlev NA, Bartek J, Bazan NG, Becker C, Bernassola F, Bertrand MJM, Bianchi ME, Blagosklonny MV, Blander JM, Blandino G, Blomgren K, Borner C, Bortner CD, Bove P, Boya P, Brenner C, Broz P, Brunner T, Damgaard RB, Calin GA, Campanella M, Candi E, Carbone M, Carmona-Gutierrez D, Cecconi F, Chan FK, Chen GQ, Chen Q, Chen YH, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Ciliberto G, Conrad M, Cubillos-Ruiz JR, Czabotar PE, D'Angiolella V, Daugaard M, Dawson TM, Dawson VL, De Maria R, De Strooper B, Debatin KM, Deberardinis RJ, Degterev A, Del Sal G, Deshmukh M, Di Virgilio F, Diederich M, Dixon SJ, Dynlacht BD, El-Deiry WS, Elrod JW, Engeland K, Fimia GM, Galassi C, Ganini C, Garcia-Saez AJ, Garg AD, Garrido C, Gavathiotis E, Gerlic M, Ghosh S, Green DR, Greene LA, Gronemeyer H, Häcker G, Hajnóczky G, Hardwick JM, Haupt Y, He S, Heery DM, Hengartner MO, Hetz C, Hildeman DA, Ichijo H, Inoue S, Jäättelä M, Janic A, Joseph B, Jost PJ, Kanneganti TD, Karin M, Kashkar H, Kaufmann T, Kelly GL, Kepp O, Kimchi A, Kitsis RN, Klionsky DJ, Kluck R, Krysko DV, Kulms D, Kumar S, Lavandero S, Lavrik IN, Lemasters JJ, Liccardi G, Linkermann A, Lipton SA, Lockshin RA, López-Otín C, Luedde T, MacFarlane M, Madeo F, Malorni W, Manic G, Mantovani R, Marchi S, Marine JC, Martin SJ, Martinou JC, Mastroberardino PG, Medema JP, Mehlen P, Meier P, Melino G, Melino S, Miao EA, Moll UM, Muñoz-Pinedo C, Murphy DJ, Niklison-Chirou MV, Novelli F, Núñez G, Oberst A, Ofengeim D, Opferman JT, Oren M, Pagano M, Panaretakis T, Pasparakis M, Penninger JM, Pentimalli F, Pereira DM, Pervaiz S, Peter ME, Pinton P, Porta G, Prehn JHM, Puthalakath H, Rabinovich GA, Rajalingam K, Ravichandran KS, Rehm M, Ricci JE, Rizzuto R, Robinson N, Rodrigues CMP, Rotblat B, Rothlin CV, Rubinsztein DC, Rudel T, Rufini A, Ryan KM, Sarosiek KA, Sawa A, Sayan E, Schroder K, Scorrano L, Sesti F, Shao F, Shi Y, Sica GS, Silke J, Simon HU, Sistigu A, Stephanou A, Stockwell BR, Strapazzon F, Strasser A, Sun L, Sun E, Sun Q, Szabadkai G, Tait SWG, Tang D, Tavernarakis N, Troy CM, Turk B, Urbano N, Vandenabeele P, Vanden Berghe T, Vander Heiden MG, Vanderluit JL, Verkhratsky A, Villunger A, von Karstedt S, Voss AK, Vousden KH, Vucic D, Vuri D, Wagner EF, Walczak H, Wallach D, Wang R, Wang Y, Weber A, Wood W, Yamazaki T, Yang HT, Zakeri Z, Zawacka-Pankau JE, Zhang L, Zhang H, Zhivotovsky B, Zhou W, Piacentini M, Kroemer G, and Galluzzi L
- Subjects
- Animals, Humans, Cell Death, Carcinogenesis, Mammals metabolism, Apoptosis genetics, Caspases genetics, Caspases metabolism
- Abstract
Apoptosis is a form of regulated cell death (RCD) that involves proteases of the caspase family. Pharmacological and genetic strategies that experimentally inhibit or delay apoptosis in mammalian systems have elucidated the key contribution of this process not only to (post-)embryonic development and adult tissue homeostasis, but also to the etiology of multiple human disorders. Consistent with this notion, while defects in the molecular machinery for apoptotic cell death impair organismal development and promote oncogenesis, the unwarranted activation of apoptosis promotes cell loss and tissue damage in the context of various neurological, cardiovascular, renal, hepatic, infectious, neoplastic and inflammatory conditions. Here, the Nomenclature Committee on Cell Death (NCCD) gathered to critically summarize an abundant pre-clinical literature mechanistically linking the core apoptotic apparatus to organismal homeostasis in the context of disease., (© 2023. The Author(s), under exclusive licence to ADMC Associazione Differenziamento e Morte Cellulare.)
- Published
- 2023
- Full Text
- View/download PDF
13. DNA methylation of the promoter region at the CREB1 binding site is a mechanism for the epigenetic regulation of brain-specific PKMζ.
- Author
-
Pramio DT, Vieceli FM, Varella-Branco E, Goes CP, Kobayashi GS, da Silva Pelegrina DV, de Moraes BC, El Allam A, De Kumar B, Jara G, Farfel JM, Bennett DA, Kundu S, Viapiano MS, Reis EM, de Oliveira PSL, Dos Santos E Passos-Bueno MR, Rothlin CV, Ghosh S, and Schechtman D
- Subjects
- Humans, DNA Methylation, Epigenesis, Genetic, Long-Term Potentiation physiology, Brain, Cyclic AMP Response Element-Binding Protein genetics, Alzheimer Disease
- Abstract
Protein kinase M zeta, PKMζ, is a brain enriched kinase with a well characterized role in Long-Term Potentiation (LTP), the activity-dependent strengthening of synapses involved in long-term memory formation. However, little is known about the molecular mechanisms that maintain the tissue specificity of this kinase. Here, we characterized the epigenetic factors, mainly DNA methylation, regulating PKMζ expression in the human brain. The PRKCZ gene has an upstream promoter regulating Protein kinase C ζ (PKCζ), and an internal promoter driving PKMζ expression. A demethylated region, including a canonical CREB binding site, situated at the internal promoter was only observed in human CNS tissues. The induction of site-specific hypermethylation of this region resulted in decreased CREB1 binding and downregulation of PKMζ expression. Noteworthy, CREB binding sites were absent in the upstream promoter of PRKCZ locus, suggesting a specific mechanism for regulating PKMζ expression. These observations were validated using a system of human neuronal differentiation from induced pluripotent stem cells (iPSCs). CREB1 binding at the internal promoter was detected only in differentiated neurons, where PKMζ is expressed. The same epigenetic mechanism in the context of CREB binding site was identified in other genes involved in neuronal differentiation and LTP. Additionally, aberrant DNA hypermethylation at the internal promoter was observed in cases of Alzheimer's disease, correlating with decreased expression of PKMζ in patient brains. Altogether, we present a conserved epigenetic mechanism regulating PKMζ expression and other genes enhanced in the CNS with possible implications in neuronal differentiation and Alzheimer's disease., Competing Interests: Declaration of competing interest The authors no conflict of interest., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
14. Single-cell transcriptomic atlas of Alzheimer's disease middle temporal gyrus reveals region, cell type and sex specificity of gene expression with novel genetic risk for MERTK in female.
- Author
-
Zhang L, He CH, Coffey S, Yin D, Hsu IU, Su C, Ye Y, Zhang C, Spurrier J, Nicholson L, Rothlin CV, Ghosh S, Gopal PP, Hafler DA, Zhao H, and Strittmatter SM
- Abstract
Alzheimer's disease, the most common age-related neurodegenerative disease, is closely associated with both amyloid-ß plaque and neuroinflammation. Two thirds of Alzheimer's disease patients are females and they have a higher disease risk. Moreover, women with Alzheimer's disease have more extensive brain histological changes than men along with more severe cognitive symptoms and neurodegeneration. To identify how sex difference induces structural brain changes, we performed unbiased massively parallel single nucleus RNA sequencing on Alzheimer's disease and control brains focusing on the middle temporal gyrus, a brain region strongly affected by the disease but not previously studied with these methods. We identified a subpopulation of selectively vulnerable layer 2/3 excitatory neurons that that were RORB-negative and CDH9-expressing. This vulnerability differs from that reported for other brain regions, but there was no detectable difference between male and female patterns in middle temporal gyrus samples. Disease-associated, but sex-independent, reactive astrocyte signatures were also present. In clear contrast, the microglia signatures of diseased brains differed between males and females. Combining single cell transcriptomic data with results from genome-wide association studies (GWAS), we identified MERTK genetic variation as a risk factor for Alzheimer's disease selectively in females. Taken together, our single cell dataset revealed a unique cellular-level view of sex-specific transcriptional changes in Alzheimer's disease, illuminating GWAS identification of sex-specific Alzheimer's risk genes. These data serve as a rich resource for interrogation of the molecular and cellular basis of Alzheimer's disease.
- Published
- 2023
- Full Text
- View/download PDF
15. When aging gets on the way of disposal: Senescent cell suppression of efferocytosis.
- Author
-
Rothlin CV and Ghosh S
- Subjects
- Humans, Inflammation, Aging, Cellular Senescence, Macrophages, Phagocytosis
- Abstract
Chronic senescence can trigger pathological inflammation. In this issue, Schloesser et al. (2022. J. Cell Biol.https://doi.org/10.1083/jcb.202207097) demonstrate that senescent cells employ "don't eat me" signals that inhibit the ability of macrophages to engulf them and additionally prevent macrophages from removing neighboring corpses, revealing a new mechanism by which senescence may contribute to triggering inflammation., (© 2023 Rothlin and Ghosh.)
- Published
- 2023
- Full Text
- View/download PDF
16. Inflammation of the retinal pigment epithelium drives early-onset photoreceptor degeneration in Mertk -associated retinitis pigmentosa.
- Author
-
Mercau ME, Akalu YT, Mazzoni F, Gyimesi G, Alberto EJ, Kong Y, Hafler BP, Finnemann SC, Rothlin CV, and Ghosh S
- Subjects
- Mice, Animals, c-Mer Tyrosine Kinase genetics, c-Mer Tyrosine Kinase metabolism, Retinal Pigment Epithelium metabolism, Receptor Protein-Tyrosine Kinases genetics, Receptor Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Inflammation genetics, Inflammation metabolism, Retinal Degeneration genetics, Retinal Degeneration metabolism, Retinitis Pigmentosa genetics, Retinitis Pigmentosa metabolism
- Abstract
Severe, early-onset photoreceptor (PR) degeneration associated with MERTK mutations is thought to result from failed phagocytosis by retinal pigment epithelium (RPE). Notwithstanding, the severity and onset of PR degeneration in mouse models of Mertk ablation are determined by the hypomorphic expression or the loss of the Mertk paralog Tyro3 . Here, we find that loss of Mertk and reduced expression/loss of Tyro3 led to RPE inflammation even before eye-opening. Incipient RPE inflammation cascaded to involve microglia activation and PR degeneration with monocyte infiltration. Inhibition of RPE inflammation with the JAK1/2 inhibitor ruxolitinib mitigated PR degeneration in Mertk
-/- mice. Neither inflammation nor severe, early-onset PR degeneration was observed in mice with defective phagocytosis alone. Thus, inflammation drives severe, early-onset PR degeneration-associated with Mertk loss of function.- Published
- 2023
- Full Text
- View/download PDF
17. Cell death in development, maintenance, and diseases of the nervous system.
- Author
-
Mercau ME, Patwa S, Bhat KPL, Ghosh S, and Rothlin CV
- Subjects
- Adult, Cell Death, Homeostasis, Humans, Neurons, Nervous System, Neurogenesis physiology
- Abstract
Cell death, be it of neurons or glial cells, marks the development of the nervous system. Albeit relatively less so than in tissues such as the gut, cell death is also a feature of nervous system homeostasis-especially in context of adult neurogenesis. Finally, cell death is commonplace in acute brain injuries, chronic neurodegenerative diseases, and in some central nervous system tumors such as glioblastoma. Recent studies are enumerating the various molecular modalities involved in the execution of cells. Intimately linked with cell death are mechanisms of disposal that remove the dead cell and bring about a tissue-level response. Heretofore, the association between these methods of dying and physiological or pathological responses has remained nebulous. It is envisioned that careful cartography of death and disposal may reveal novel understandings of disease states and chart new therapeutic strategies in the near future., (© 2022. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
18. Tissue-specific modifier alleles determine Mertk loss-of-function traits.
- Author
-
Akalu YT, Mercau ME, Ansems M, Hughes LD, Nevin J, Alberto EJ, Liu XN, He LZ, Alvarado D, Keler T, Kong Y, Philbrick WM, Bosenberg M, Finnemann SC, Iavarone A, Lasorella A, Rothlin CV, and Ghosh S
- Subjects
- Alleles, Animals, Disease Models, Animal, Mice, Mice, Knockout, Phagocytosis genetics, Phenotype, Proto-Oncogene Proteins genetics, Retinal Pigments, c-Mer Tyrosine Kinase genetics, c-Mer Tyrosine Kinase metabolism, Retinal Degeneration genetics, Retinal Degeneration pathology
- Abstract
Knockout (KO) mouse models play critical roles in elucidating biological processes behind disease-associated or disease-resistant traits. As a presumed consequence of gene KO, mice display certain phenotypes. Based on insight into the molecular role of said gene in a biological process, it is inferred that the particular biological process causally underlies the trait. This approach has been crucial towards understanding the basis of pathological and/or advantageous traits associated with Mertk KO mice. Mertk KO mice suffer from severe, early-onset retinal degeneration. MERTK, expressed in retinal pigment epithelia, is a receptor tyrosine kinase with a critical role in phagocytosis of apoptotic cells or cellular debris. Therefore, early-onset, severe retinal degeneration was described to be a direct consequence of failed MERTK-mediated phagocytosis of photoreceptor outer segments by retinal pigment epithelia. Here, we report that the loss of Mertk alone is not sufficient for retinal degeneration. The widely used Mertk KO mouse carries multiple coincidental changes in its genome that affect the expression of a number of genes, including the Mertk paralog Tyro3 . Retinal degeneration manifests only when the function of Tyro3 is concomitantly lost. Furthermore, Mertk KO mice display improved anti-tumor immunity. MERTK is expressed in macrophages. Therefore, enhanced anti-tumor immunity was inferred to result from the failure of macrophages to dispose of cancer cell corpses, resulting in a pro-inflammatory tumor microenvironment. The resistance against two syngeneic mouse tumor models observed in Mertk KO mice is not, however, phenocopied by the loss of Mertk alone. Neither Tyro3 nor macrophage phagocytosis by alternate genetic redundancy accounts for the absence of anti-tumor immunity. Collectively, our results indicate that context-dependent epistasis of independent modifier alleles determines Mertk KO traits., Competing Interests: YA, MM, MA, LH, JN, EA, XL, YK, WP, MB, SF, AI, AL No competing interests declared, LH, DA, TK is affiliated with Celldex Therapeutics. The author has no financial interests to declare, CR Senior editor, eLife, SG has received grant support from Mirati Therapeutics, (© 2022, Akalu, Mercau et al.)
- Published
- 2022
- Full Text
- View/download PDF
19. Phosphatidylserine Synthase PTDSS1 Shapes the Tumor Lipidome to Maintain Tumor-Promoting Inflammation.
- Author
-
Sekar D, Dillmann C, Sirait-Fischer E, Fink AF, Zivkovic A, Baum N, Strack E, Klatt S, Zukunft S, Wallner S, Descot A, Olesch C, da Silva P, von Knethen A, Schmid T, Grösch S, Savai R, Ferreirós N, Fleming I, Ghosh S, Rothlin CV, Stark H, Medyouf H, Brüne B, and Weigert A
- Subjects
- Animals, CDPdiacylglycerol-Serine O-Phosphatidyltransferase, Ether, Humans, Inflammation metabolism, Mice, Phosphatidylserines metabolism, Tumor Microenvironment, c-Mer Tyrosine Kinase metabolism, Lipidomics, Neoplasms metabolism
- Abstract
An altered lipidome in tumors may affect not only tumor cells themselves but also their microenvironment. In this study, a lipidomics screen reveals increased amounts of phosphatidylserine (PS), particularly ether-PS (ePS), in murine mammary tumors compared with normal tissue. PS was produced by phosphatidylserine synthase 1 (PTDSS1), and depletion of Ptdss1 from tumor cells in mice reduced ePS levels accompanied by stunted tumor growth and decreased tumor-associated macrophage (TAM) abundance. Ptdss1-deficient tumor cells exposed less PS during apoptosis, which was recognized by the PS receptor MERTK. Mammary tumors in macrophage-specific Mertk-/- mice showed similarly suppressed growth and reduced TAM infiltration. Transcriptomic profiles of TAMs from Ptdss1-knockdown tumors and Mertk-/- TAMs revealed that macrophage proliferation was reduced when the Ptdss1/Mertk pathway was targeted. Moreover, PTDSS1 expression correlated positively with TAM abundance but negatively with breast carcinoma patient survival. PTDSS1 thus may be a target to modify tumor-promoting inflammation., Significance: This study shows that inhibiting the production of ether-phosphatidylserine by targeting phosphatidylserine synthase PTDSS1 limits tumor-associated macrophage expansion and breast tumor growth., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
20. The purinergic P2Y14 receptor links hepatocyte death to hepatic stellate cell activation and fibrogenesis in the liver.
- Author
-
Mederacke I, Filliol A, Affo S, Nair A, Hernandez C, Sun Q, Hamberger F, Brundu F, Chen Y, Ravichandra A, Huebener P, Anke H, Shi H, Martínez García de la Torre RA, Smith JR, Henderson NC, Vondran FWR, Rothlin CV, Baehre H, Tabas I, Sancho-Bru P, and Schwabe RF
- Subjects
- Animals, Extracellular Signal-Regulated MAP Kinases metabolism, Humans, Ligands, Liver metabolism, Liver Cirrhosis pathology, Mice, Proteomics, Single-Cell Analysis, Uridine Diphosphate metabolism, YAP-Signaling Proteins, Hepatic Stellate Cells metabolism, Hepatocytes metabolism, Receptors, Purinergic P2 metabolism, Receptors, Purinergic P2Y metabolism
- Abstract
Fibrosis contributes to ~45% of deaths in western countries. In chronic liver disease, fibrosis is a major factor determining outcomes, but efficient antifibrotic therapies are lacking. Although platelet-derived growth factor and transforming growth factor-β constitute key fibrogenic mediators, they do not account for the well-established link between cell death and fibrosis in the liver. Here, we hypothesized that damage-associated molecular patterns (DAMPs) may link epithelial cell death to fibrogenesis in the injured liver. DAMP receptor screening identified purinergic receptor P2Y14 among several candidates as highly enriched in hepatic stellate cells (HSCs), the main fibrogenic cell type of the liver. Conversely, P2Y14 ligands uridine 5'-diphosphate (UDP)-glucose and UDP-galactose were enriched in hepatocytes and were released upon different modes of cell death. Accordingly, ligand-receptor interaction analysis that combined proteomic and single-cell RNA sequencing data revealed P2Y14 ligands and P2Y14 receptor as a link between dying cells and HSCs, respectively. Treatment with P2Y14 ligands or coculture with dying hepatocytes promoted HSC activation in a P2Y14-dependent manner. P2Y14 ligands activated extracellular signal-regulated kinase (ERK) and Yes-associated protein (YAP) signaling in HSCs, resulting in ERK-dependent HSC activation. Global and HSC-selective P2Y14 deficiency attenuated liver fibrosis in multiple mouse models of liver injury. Functional expression of P2Y14 was confirmed in healthy and diseased human liver and human HSCs. In conclusion, P2Y14 ligands and their receptor constitute a profibrogenic DAMP pathway that directly links cell death to fibrogenesis.
- Published
- 2022
- Full Text
- View/download PDF
21. Inhibition of renalase drives tumour rejection by promoting T cell activation.
- Author
-
Guo X, Jessel S, Qu R, Kluger Y, Chen TM, Hollander L, Safirstein R, Nelson B, Cha C, Bosenberg M, Jilaveanu LB, Rimm D, Rothlin CV, Kluger HM, and Desir GV
- Subjects
- Animals, Humans, Immunotherapy, Mice, Monoamine Oxidase therapeutic use, Tumor Microenvironment, Immune Checkpoint Inhibitors, Melanoma drug therapy
- Abstract
Background: Although programmed cell death protein 1 (PD-1) inhibitors have revolutionised treatment for advanced melanoma, not all patients respond. We previously showed that inhibition of the flavoprotein renalase (RNLS) in preclinical melanoma models decreases tumour growth. We hypothesised that RNLS inhibition promotes tumour rejection by effects on the tumour microenvironment (TME)., Methods: We used two distinct murine melanoma models, studied in RNLS knockout (KO) or wild-type (WT) mice. WT mice were treated with the anti-RNLS antibody, m28, with or without anti-PD-1. 10X single-cell RNA-sequencing was used to identify transcriptional differences between treatment groups, and tumour cell content was interrogated by flow cytometry. Samples from patients treated with immunotherapy were examined for RNLS expression by quantitative immunofluorescence., Results: RNLS KO mice injected with wild-type melanoma cells reject their tumours, supporting the importance of RNLS in cells in the TME. This effect was blunted by anti-cluster of differentiation 3. However, MØ-specific RNLS ablation was insufficient to abrogate tumour formation. Anti-RNLS antibody treatment of melanoma-bearing mice resulted in enhanced T cell infiltration and activation and resulted in immune memory on rechallenging mice with injection of melanoma cells. At the single-cell level, treatment with anti-RNLS antibodies resulted in increased tumour density of MØ, neutrophils and lymphocytes and increased expression of IFNγ and granzyme B in natural killer cells and T cells. Intratumoural Forkhead Box P3 + CD4 cells were decreased. In two distinct murine melanoma models, we showed that melanoma-bearing mice treated with anti-RNLS antibodies plus anti-PD-1 had superior tumour shrinkage and survival than with either treatment alone. Importantly, in pretreatment samples from patients treated with PD-1 inhibitors, high RNLS expression was associated with decreased survival (log-rank P = 0.006), independent of other prognostic variables., Conclusions: RNLS KO results in melanoma tumour regression in a T-cell-dependent fashion. Anti-RNLS antibodies enhance anti-PD-1 activity in two distinct aggressive murine melanoma models resistant to PD-1 inhibitors, supporting the development of anti-RNLS antibodies with PD-1 inhibitors as a novel approach for melanomas poorly responsive to anti-PD-1., Competing Interests: Conflict of interest statement The authors declare the following financial interests/personal relationships which may be considered potential competing interests: G Desir is a named inventor on several issued patents related to the discovery and therapeutic use of renalase. Renalase is licensed to Bessor Pharma, and G Desir holds an equity position in Bessor and its subsidiary Personal Therapeutics. H. Kluger has received institutional research grants from Merck, Bristol-Myers Squibb and Apexigen; personal fees from Nektar, Iovance, Immunocore, Celldex, Array Biopharma, Merck, Elevate Bio, Instil Bio, Bristol-Myers Squibb, Clinigen, Shionogi, Chemocentryx and Calithera. All are outside of the reported results. All remaining authors have declared no conflicts of interest., (Copyright © 2022 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
22. AXL Inhibition in Macrophages Stimulates Host-versus-Leukemia Immunity and Eradicates Naïve and Treatment-Resistant Leukemia.
- Author
-
Tirado-Gonzalez I, Descot A, Soetopo D, Nevmerzhitskaya A, Schäffer A, Kur IM, Czlonka E, Wachtel C, Tsoukala I, Müller L, Schäfer AL, Weitmann M, Dinse P, Alberto E, Buck MC, Landry JJ, Baying B, Slotta-Huspenina J, Roesler J, Harter PN, Kubasch AS, Meinel J, Elwakeel E, Strack E, Quang CT, Abdel-Wahab O, Schmitz M, Weigert A, Schmid T, Platzbecker U, Benes V, Ghysdael J, Bonig H, Götze KS, Rothlin CV, Ghosh S, and Medyouf H
- Subjects
- Humans, Immunotherapy, Macrophages, Signal Transduction, Leukemia therapy
- Abstract
Acute leukemias are systemic malignancies associated with a dire outcome. Because of low immunogenicity, leukemias display a remarkable ability to evade immune control and are often resistant to checkpoint blockade. Here, we discover that leukemia cells actively establish a suppressive environment to prevent immune attacks by co-opting a signaling axis that skews macrophages toward a tumor-promoting tissue repair phenotype, namely the GAS6/AXL axis. Using aggressive leukemia models, we demonstrate that ablation of the AXL receptor specifically in macrophages, or its ligand GAS6 in the environment, stimulates antileukemic immunity and elicits effective and lasting natural killer cell- and T cell-dependent immune response against naïve and treatment-resistant leukemia. Remarkably, AXL deficiency in macrophages also enables PD-1 checkpoint blockade in PD-1-refractory leukemias. Finally, we provide proof-of-concept that a clinical-grade AXL inhibitor can be used in combination with standard-of-care therapy to cure established leukemia, regardless of AXL expression in malignant cells. SIGNIFICANCE: Alternatively primed myeloid cells predict negative outcome in leukemia. By demonstrating that leukemia cells actively evade immune control by engaging AXL receptor tyrosine kinase in macrophages and promoting their alternative priming, we identified a target which blockade, using a clinical-grade inhibitor, is vital to unleashing the therapeutic potential of myeloid-centered immunotherapy. This article is highlighted in the In This Issue feature, p. 2659 ., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
23. Gas6 drives Zika virus-induced neurological complications in humans and congenital syndrome in immunocompetent mice.
- Author
-
Silva-Filho JL, de Oliveira LG, Monteiro L, Parise PL, Zanluqui NG, Polonio CM, de Freitas CL, Toledo-Teixeira DA, de Souza WM, Bittencourt N, Amorim MR, Forato J, Muraro SP, de Souza GF, Martini MC, Bispo-Dos-Santos K, Vieira A, Judice CC, Pastore GM, Amaral E, Passini Junior R, Mayer-Milanez HMBP, Ribeiro-do-Valle CC, Calil R, Renato Bennini Junior J, Lajos GJ, Altemani A, Nolasco da Silva MT, Carolina Coan A, Francisca Colella-Santos M, von Zuben APB, Vinolo MAR, Arns CW, Catharino RR, Costa ML, Angerami RN, Freitas ARR, Resende MR, Garcia MT, Luiza Moretti M, Renia L, Ng LFP, Rothlin CV, Costa FTM, Peron JPS, and Proença-Modena JL
- Subjects
- Animals, Female, Humans, Mice, Placenta, Pregnancy, Virus Replication, Nervous System Diseases, Zika Virus, Zika Virus Infection complications
- Abstract
Zika virus (ZIKV) has the ability to cross placental and brain barriers, causing congenital malformations in neonates and neurological disorders in adults. However, the pathogenic mechanisms of ZIKV-induced neurological complications in adults and congenital malformations are still not fully understood. Gas6 is a soluble TAM receptor ligand able to promote flavivirus internalization and downregulation of immune responses. Here we demonstrate that there is a correlation between ZIKV neurological complications with higher Gas6 levels and the downregulation of genes associated with anti-viral response, as type I IFN due to Socs1 upregulation. Also, Gas6 gamma-carboxylation is essential for ZIKV invasion and replication in monocytes, the main source of this protein, which was inhibited by warfarin. Conversely, Gas6 facilitates ZIKV replication in adult immunocompetent mice and enabled susceptibility to transplacental infection. Our data indicate that ZIKV promotes the upregulation of its ligand Gas6, which contributes to viral infectivity and drives the development of severe adverse outcomes during ZIKV infection., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
24. Efferocytosis fuels malignant pleural effusion through TIMP1.
- Author
-
Zhao L, Giannou AD, Xu Y, Shiri AM, Liebold I, Steglich B, Bedke T, Zhang T, Lücke J, Scognamiglio P, Kempski J, Woestemeier A, Chen J, Agalioti T, Zazara DE, Lindner D, Janning M, Hennigs JK, Jagirdar RM, Kotsiou OS, Zarogiannis SG, Kobayashi Y, Izbicki JR, Ghosh S, Rothlin CV, Bosurgi L, Huber S, and Gagliani N
- Abstract
Malignant pleural effusion (MPE) results from the capacity of several human cancers to metastasize to the pleural cavity. No effective treatments are currently available, reflecting our insufficient understanding of the basic mechanisms leading to MPE progression. Here, we found that efferocytosis through the receptor tyrosine kinases AXL and MERTK led to the production of interleukin-10 (IL-10) by four distinct pleural cavity macrophage (Mφ) subpopulations characterized by different metabolic states and cell chemotaxis properties. In turn, IL-10 acts on dendritic cells (DCs) inducing the production of tissue inhibitor of metalloproteinases 1 (TIMP1). Genetic ablation of Axl and Mertk in Mφs or IL-10 receptor in DCs or Timp1 substantially reduced MPE progression. Our results delineate an inflammatory cascade-from the clearance of apoptotic cells by Mφs, to production of IL-10, to induction of TIMP1 in DCs-that facilitates MPE progression. This inflammatory cascade offers a series of therapeutic targets for MPE., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2021
- Full Text
- View/download PDF
25. Management of cell death in parasitic infections.
- Author
-
Bosurgi L and Rothlin CV
- Subjects
- Animals, Apoptosis, Humans, Macrophages, Phosphatidylserines, Parasites, Parasitic Diseases
- Abstract
For a long time, host cell death during parasitic infection has been considered a reflection of tissue damage, and often associated with disease pathogenesis. However, during their evolution, protozoan and helminth parasites have developed strategies to interfere with cell death so as to spread and survive in the infected host, thereby ascribing a more intriguing role to infection-associated cell death. In this review, we examine the mechanisms used by intracellular and extracellular parasites to respectively inhibit or trigger programmed cell death. We further dissect the role of the prototypical "eat-me signal" phosphatidylserine (PtdSer) which, by being exposed on the cell surface of damaged host cells as well as on some viable parasites via a process of apoptotic mimicry, leads to their recognition and up-take by the neighboring phagocytes. Although barely dissected so far, the engagement of different PtdSer receptors on macrophages, by shaping the host immune response, affects the overall infection outcome in models of both protozoan and helminth infections. In this scenario, further understanding of the molecular and cellular regulation of the PtdSer exposing cell-macrophage interaction might allow the identification of new therapeutic targets for the management of parasitic infection., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
26. Targeting Axl favors an antitumorigenic microenvironment that enhances immunotherapy responses by decreasing Hif-1α levels.
- Author
-
Goyette MA, Elkholi IE, Apcher C, Kuasne H, Rothlin CV, Muller WJ, Richard DE, Park M, Gratton JP, and Côté JF
- Subjects
- Animals, Breast Neoplasms drug therapy, Breast Neoplasms physiopathology, Cell Line, Tumor, Cell Proliferation drug effects, Epithelial-Mesenchymal Transition drug effects, Female, Gene Deletion, Gene Expression Regulation, Neoplastic drug effects, Gene Targeting, Humans, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Immune Checkpoint Inhibitors administration & dosage, Macrophages drug effects, Macrophages immunology, Mice, Neoplasm Metastasis drug therapy, Neoplasm Metastasis genetics, Neoplasm Metastasis immunology, Programmed Cell Death 1 Receptor genetics, Programmed Cell Death 1 Receptor immunology, Proto-Oncogene Proteins immunology, Receptor Protein-Tyrosine Kinases immunology, Tumor Microenvironment drug effects, Axl Receptor Tyrosine Kinase, Breast Neoplasms genetics, Breast Neoplasms immunology, Hypoxia-Inducible Factor 1, alpha Subunit immunology, Immunotherapy, Proto-Oncogene Proteins genetics, Receptor Protein-Tyrosine Kinases genetics
- Abstract
Hypoxia is an important phenomenon in solid tumors that contributes to metastasis, tumor microenvironment (TME) deregulation, and resistance to therapies. The receptor tyrosine kinase AXL is an HIF target, but its roles during hypoxic stress leading to the TME deregulation are not well defined. We report here that the mammary gland-specific deletion of Axl in a HER2
+ mouse model of breast cancer leads to a normalization of the blood vessels, a proinflammatory TME, and a reduction of lung metastases by dampening the hypoxic response in tumor cells. During hypoxia, interfering with AXL reduces HIF-1α levels altering the hypoxic response leading to a reduction of hypoxia-induced epithelial-to-mesenchymal transition (EMT), invasion, and production of key cytokines for macrophages behaviors. These observations suggest that inhibition of Axl generates a suitable setting to increase immunotherapy. Accordingly, combining pharmacological inhibition of Axl with anti-PD-1 in a preclinical model of HER2+ breast cancer reduces the primary tumor and metastatic burdens, suggesting a potential therapeutic approach to manage HER2+ patients whose tumors present high hypoxic features., Competing Interests: The authors declare no competing interest., (Copyright © 2021 the Author(s). Published by PNAS.)- Published
- 2021
- Full Text
- View/download PDF
27. Ligand-dependent kinase activity of MERTK drives efferocytosis in human iPSC-derived macrophages.
- Author
-
Wanke F, Gutbier S, Rümmelin A, Steinberg M, Hughes LD, Koenen M, Komuczki J, Regan-Komito D, Wagage S, Hesselmann J, Thoma R, Brugger D, Christopeit T, Wang H, Point F, Hallet R, Ghosh S, Rothlin CV, Patsch C, and Geering B
- Subjects
- Animals, Cell Differentiation, Cells, Cultured, HEK293 Cells, Humans, Induced Pluripotent Stem Cells drug effects, Induced Pluripotent Stem Cells physiology, Intercellular Signaling Peptides and Proteins physiology, Ligands, Macrophages drug effects, Mice, Mice, Knockout, Phagocytosis drug effects, Phagocytosis genetics, Phosphatidylserines pharmacology, c-Mer Tyrosine Kinase agonists, c-Mer Tyrosine Kinase genetics, Macrophages physiology, Phagocytosis physiology, c-Mer Tyrosine Kinase metabolism
- Abstract
Removal of apoptotic cells by phagocytes (also called efferocytosis) is a crucial process for tissue homeostasis. Professional phagocytes express a plethora of surface receptors enabling them to sense and engulf apoptotic cells, thus avoiding persistence of dead cells and cellular debris and their consequent effects. Dysregulation of efferocytosis is thought to lead to secondary necrosis and associated inflammation and immune activation. Efferocytosis in primarily murine macrophages and dendritic cells has been shown to require TAM RTKs, with MERTK and AXL being critical for clearance of apoptotic cells. The functional role of human orthologs, especially the exact contribution of each individual receptor is less well studied. Here we show that human macrophages differentiated in vitro from iPSC-derived precursor cells express both AXL and MERTK and engulf apoptotic cells. TAM RTK agonism by the natural ligand growth-arrest specific 6 (GAS6) significantly enhanced such efferocytosis. Using a newly-developed mouse model of kinase-dead MERTK, we demonstrate that MERTK kinase activity is essential for efferocytosis in peritoneal macrophages in vivo. Moreover, human iPSC-derived macrophages treated in vitro with blocking antibodies or small molecule inhibitors recapitulated this observation. Hence, our results highlight a conserved MERTK function between mice and humans, and the critical role of its kinase activity in homeostatic efferocytosis.
- Published
- 2021
- Full Text
- View/download PDF
28. Determining the effector response to cell death.
- Author
-
Rothlin CV, Hille TD, and Ghosh S
- Subjects
- Animals, Cellular Microenvironment immunology, Cytokines immunology, Homeostasis immunology, Host-Pathogen Interactions immunology, Humans, Immunity, Innate, Mathematical Concepts, Models, Immunological, Phagocytes classification, Phagocytes immunology, Phagocytosis immunology, Signal Transduction immunology, Cell Death immunology
- Abstract
Cell death occurs when a pathogen invades a host organism or the organism is subjected to sterile injury. Thus, cell death is often closely associated with the induction of an immune response. Furthermore, cell death can occur as a consequence of the immune response and precedes the tissue renewal and repair responses that are initiated by innate immune cells during resolution of an immune response. Beyond immunity, cell death is required for development, morphogenesis and homeostasis. How can such a ubiquitous event as cell death trigger such a wide range of context-specific effector responses? Dying cells are sensed by innate immune cells using specialized receptors and phagocytosed through a process termed efferocytosis. Here, we outline a general principle whereby signals within the dead cell as well as the environment are integrated by specific efferocytes to define the appropriate effector response.
- Published
- 2021
- Full Text
- View/download PDF
29. Decoding Cell Death: From a Veritable Library of Babel to Vade Mecum ?
- Author
-
Hughes LD, Wang Y, Meli AP, Rothlin CV, and Ghosh S
- Subjects
- Animals, Cell Death, Homeostasis, Humans, Apoptosis
- Abstract
Programmed cell death (PCD) is a requisite feature of development and homeostasis but can also be indicative of infections, injuries, and pathologies. In concordance with these heterogeneous contexts, an array of disparate effector responses occur downstream of cell death and its clearance-spanning tissue morphogenesis, homeostatic turnover, host defense, active dampening of inflammation, and tissue repair. This raises a fundamental question of how a single contextually appropriate response ensues after an event of PCD. To explore how complex inputs may together tailor the specificity of the resulting effector response, here we consider ( a ) the varying contexts during which different cell death modalities are observed, ( b ) the nature of the information that can be passed on by cell corpses, and ( c ) the ways by which efferocyte populations synthesize signals from dying cells with those from the surrounding microenvironment.
- Published
- 2021
- Full Text
- View/download PDF
30. Macrophage AXL receptor tyrosine kinase inflames the heart after reperfused myocardial infarction.
- Author
-
DeBerge M, Glinton K, Subramanian M, Wilsbacher LD, Rothlin CV, Tabas I, and Thorp EB
- Subjects
- Animals, Disease Models, Animal, Female, Humans, Inflammasomes metabolism, Macrophage Activation, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocardial Reperfusion Injury complications, Myocardial Reperfusion Injury metabolism, Proto-Oncogene Proteins deficiency, Proto-Oncogene Proteins genetics, Receptor Cross-Talk, Receptor Protein-Tyrosine Kinases deficiency, Receptor Protein-Tyrosine Kinases genetics, ST Elevation Myocardial Infarction metabolism, Signal Transduction, Toll-Like Receptor 4 metabolism, c-Mer Tyrosine Kinase deficiency, c-Mer Tyrosine Kinase genetics, c-Mer Tyrosine Kinase metabolism, Axl Receptor Tyrosine Kinase, Macrophages metabolism, Myocardial Infarction complications, Myocardial Infarction metabolism, Myocarditis etiology, Myocarditis metabolism, Proto-Oncogene Proteins metabolism, Receptor Protein-Tyrosine Kinases metabolism
- Abstract
Tyro3, AXL, and MerTK (TAM) receptors are activated in macrophages in response to tissue injury and as such have been proposed as therapeutic targets to promote inflammation resolution during sterile wound healing, including myocardial infarction. Although the role of MerTK in cardioprotection is well characterized, the unique role of the other structurally similar TAMs, and particularly AXL, in clinically relevant models of myocardial ischemia/reperfusion infarction (IRI) is comparatively unknown. Utilizing complementary approaches, validated by flow cytometric analysis of human and murine macrophage subsets and conditional genetic loss and gain of function, we uncover a maladaptive role for myeloid AXL during IRI in the heart. Cross signaling between AXL and TLR4 in cardiac macrophages directed a switch to glycolytic metabolism and secretion of proinflammatory IL-1β, leading to increased intramyocardial inflammation, adverse ventricular remodeling, and impaired contractile function. AXL functioned independently of cardioprotective MerTK to reduce the efficacy of cardiac repair, but like MerTK, was proteolytically cleaved. Administration of a selective small molecule AXL inhibitor alone improved cardiac healing, which was further enhanced in combination with blockade of MerTK cleavage. These data support further exploration of macrophage TAM receptors as therapeutic targets for myocardial infarction.
- Published
- 2021
- Full Text
- View/download PDF
31. Editorial overview: Innate immunity, from host defense and beyond.
- Author
-
Rothlin CV, Rathinam VA, and Ghosh S
- Subjects
- Humans, Host-Pathogen Interactions immunology, Immunity, Innate immunology
- Published
- 2021
- Full Text
- View/download PDF
32. GAS6 signaling tempers Th17 development in patients with multiple sclerosis and helminth infection.
- Author
-
Ortiz Wilczyñski JM, Olexen CM, Errasti AE, Schattner M, Rothlin CV, Correale J, and Carrera Silva EA
- Subjects
- Adult, Animals, Female, Humans, Male, Helminthiasis complications, Helminthiasis immunology, Intercellular Signaling Peptides and Proteins immunology, Multiple Sclerosis, Relapsing-Remitting immunology, Multiple Sclerosis, Relapsing-Remitting parasitology, Th17 Cells immunology
- Abstract
Multiple sclerosis (MS) is a highly disabling neurodegenerative autoimmune condition in which an unbalanced immune response plays a critical role. Although the mechanisms remain poorly defined, helminth infections are known to modulate the severity and progression of chronic inflammatory diseases. The tyrosine kinase receptors TYRO3, AXL, and MERTK (TAM) have been described as inhibitors of the immune response in various inflammatory settings. We show here that patients with concurrent natural helminth infections and MS condition (HIMS) had an increased expression of the negative regulatory TAM receptors in antigen-presenting cells and their agonist GAS6 in circulating CD11bhigh and CD4+ T cells compared to patients with only MS. The Th17 subset was reduced in patients with HIMS with a subsequent downregulation of its pathogenic genetic program. Moreover, these CD4+ T cells promoted lower levels of the co-stimulatory molecules CD80, CD86, and CD40 on dendritic cells compared with CD4+ T cells from patients with MS, an effect that was GAS6-dependent. IL-10+ cells from patients with HIMS showed higher GAS6 expression levels than Th17 cells, and inhibition of phosphatidylserine/GAS6 binding led to an expansion of Th17 effector genes. The addition of GAS6 on activated CD4+ T cells from patients with MS restrains the Th17 gene expression signature. This cohort of patients with HIMS unravels a promising regulatory mechanism to dampen the Th17 inflammatory response in autoimmunity., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
33. A Network of Macrophages Supports Mitochondrial Homeostasis in the Heart.
- Author
-
Nicolás-Ávila JA, Lechuga-Vieco AV, Esteban-Martínez L, Sánchez-Díaz M, Díaz-García E, Santiago DJ, Rubio-Ponce A, Li JL, Balachander A, Quintana JA, Martínez-de-Mena R, Castejón-Vega B, Pun-García A, Través PG, Bonzón-Kulichenko E, García-Marqués F, Cussó L, A-González N, González-Guerra A, Roche-Molina M, Martin-Salamanca S, Crainiciuc G, Guzmán G, Larrazabal J, Herrero-Galán E, Alegre-Cebollada J, Lemke G, Rothlin CV, Jimenez-Borreguero LJ, Reyes G, Castrillo A, Desco M, Muñoz-Cánoves P, Ibáñez B, Torres M, Ng LG, Priori SG, Bueno H, Vázquez J, Cordero MD, Bernal JA, Enríquez JA, and Hidalgo A
- Subjects
- Aged, Animals, Apoptosis, Autophagy, Female, Heart physiology, Homeostasis, Humans, Macrophages physiology, Male, Mice, Mice, Inbred C57BL, Middle Aged, Mitochondria physiology, Myocardial Infarction metabolism, Myocardium metabolism, Myocytes, Cardiac physiology, Phagocytosis physiology, Reactive Oxygen Species metabolism, Receptor Protein-Tyrosine Kinases metabolism, c-Mer Tyrosine Kinase metabolism, Macrophages metabolism, Mitochondria metabolism, Myocytes, Cardiac metabolism
- Abstract
Cardiomyocytes are subjected to the intense mechanical stress and metabolic demands of the beating heart. It is unclear whether these cells, which are long-lived and rarely renew, manage to preserve homeostasis on their own. While analyzing macrophages lodged within the healthy myocardium, we discovered that they actively took up material, including mitochondria, derived from cardiomyocytes. Cardiomyocytes ejected dysfunctional mitochondria and other cargo in dedicated membranous particles reminiscent of neural exophers, through a process driven by the cardiomyocyte's autophagy machinery that was enhanced during cardiac stress. Depletion of cardiac macrophages or deficiency in the phagocytic receptor Mertk resulted in defective elimination of mitochondria from the myocardial tissue, activation of the inflammasome, impaired autophagy, accumulation of anomalous mitochondria in cardiomyocytes, metabolic alterations, and ventricular dysfunction. Thus, we identify an immune-parenchymal pair in the murine heart that enables transfer of unfit material to preserve metabolic stability and organ function. VIDEO ABSTRACT., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
34. Astrocytes and microglia play orchestrated roles and respect phagocytic territories during neuronal corpse removal in vivo.
- Author
-
Damisah EC, Hill RA, Rai A, Chen F, Rothlin CV, Ghosh S, and Grutzendler J
- Subjects
- Cadaver, Humans, Neurons, Phagocytes, Astrocytes metabolism, Microglia
- Abstract
Cell death is prevalent throughout life; however, the coordinated interactions and roles of phagocytes during corpse removal in the live brain are poorly understood. We developed photochemical and viral methodologies to induce death in single cells and combined this with intravital optical imaging. This approach allowed us to track multicellular phagocytic interactions with precise spatiotemporal resolution. Astrocytes and microglia engaged with dying neurons in an orchestrated and synchronized fashion. Each glial cell played specialized roles: Astrocyte processes rapidly polarized and engulfed numerous small dendritic apoptotic bodies, while microglia migrated and engulfed the soma and apical dendrites. The relative involvement and phagocytic specialization of each glial cell was plastic and controlled by the receptor tyrosine kinase Mertk . In aging, there was a marked delay in apoptotic cell removal. Thus, a precisely orchestrated response and cross-talk between glial cells during corpse removal may be critical for maintaining brain homeostasis., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2020
- Full Text
- View/download PDF
35. Author Correction: A conserved dendritic-cell regulatory program limits antitumour immunity.
- Author
-
Maier B, Leader AM, Chen ST, Tung N, Chang C, LeBerichel J, Chudnovskiy A, Maskey S, Walker L, Finnigan JP, Kirkling ME, Reizis B, Ghosh S, D'Amore NR, Bhardwaj N, Rothlin CV, Wolf A, Flores R, Marron T, Rahman AH, Kenigsberg E, Brown BD, and Merad M
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
36. Cracking the Cell Death Code.
- Author
-
Rothlin CV and Ghosh S
- Subjects
- Animals, Apoptosis, Cell Differentiation, Cell Nucleus metabolism, Cell Proliferation, Cytosol metabolism, Humans, Macrophages metabolism, Models, Biological, Morphogenesis, Neurons metabolism, Cell Death physiology, Embryonic Development, Homeostasis
- Abstract
Cell death is an invariant feature throughout our life span, starting with extensive scheduled cell death during morphogenesis and continuing with death under homeostasis in adult tissues. Additionally, cells become victims of accidental, unscheduled death following injury and infection. Cell death in each of these occasions triggers specific and specialized responses in the living cells that surround them or are attracted to the dying/dead cells. These responses sculpt tissues during morphogenesis, replenish lost cells in homeostasis to maintain tissue/system function, and repair damaged tissues after injury. Wherein lies the information that sets in motion the cascade of effector responses culminating in remodeling, renewal, or repair? Here, we attempt to provide a framework for thinking about cell death in terms of the specific effector responses that accompanies various modalities of cell death. We also propose an integrated threefold "cell death code" consisting of information intrinsic to the dying/dead cell, the surroundings of the dying cell, and the identity of the responder., (Copyright © 2020 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
37. Lifting the innate immune barriers to antitumor immunity.
- Author
-
Rothlin CV and Ghosh S
- Subjects
- Humans, Immunity, Innate immunology, Immunotherapy methods, Neoplasms immunology
- Abstract
The immune system evolved for adequate surveillance and killing of pathogens while minimizing host damage, such as due to chronic or exaggerated inflammation and autoimmunity. This is achieved by negative regulators and checkpoints that limit the magnitude and time course of the immune response. Tumor cells often escape immune surveillance and killing. Therefore, disrupting the brakes built into the immune system should effectively boost the anticancer immune response. The success of anti-CTLA4, anti-PD-1 and anti-PD-L1 have firmly established this proof of concept. Since the response rate of anti-CTLA4, anti-PD-1 and anti-PD-L1 is still limited, there is an intense effort for the identification of new targets and development of approaches that can expand the benefits of immunotherapy to a larger patient pool. Additional T cell checkpoints are obvious targets; however, here we focus on the unusual suspects-cells that function to initiate and guide T cell activity. Innate immunity is both an obligate prerequisite for the initiation of adaptive immune responses and a requirement for the recruitment of activated T cells to the site of action. We discuss some of the molecules present in innate immune cells, including natural killer cells, dendritic cells, macrophages, myeloid-derived suppressor cells, endothelial cells and stromal cells, that can activate or enhance innate immune cell functions, and more importantly, the inhibitors or checkpoints present in these cells that restrain their functions. Boosting innate immunity, either by enhancing activator functions or, preferably, by blocking the inhibitors, may represent a new anticancer treatment modality or at least function as adjuvants to T cell checkpoint inhibitors., Competing Interests: Competing interests: CR is a scientific founder and member of the scientific advisory board of Surface Oncology. Work in the CR and SG laboratory has been in part supported by grants from Mirati Therapeutics., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
38. A conserved dendritic-cell regulatory program limits antitumour immunity.
- Author
-
Maier B, Leader AM, Chen ST, Tung N, Chang C, LeBerichel J, Chudnovskiy A, Maskey S, Walker L, Finnigan JP, Kirkling ME, Reizis B, Ghosh S, D'Amore NR, Bhardwaj N, Rothlin CV, Wolf A, Flores R, Marron T, Rahman AH, Kenigsberg E, Brown BD, and Merad M
- Subjects
- Animals, Antigens, Neoplasm immunology, B7-H1 Antigen immunology, B7-H1 Antigen metabolism, CD8-Positive T-Lymphocytes immunology, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung therapy, Dendritic Cells drug effects, Dendritic Cells metabolism, Humans, Immunotherapy, Interferon-gamma immunology, Interleukin-12 immunology, Interleukin-4 antagonists & inhibitors, Interleukin-4 immunology, Interleukin-4 metabolism, Lung Neoplasms pathology, Lung Neoplasms therapy, Male, Mice, Tumor Burden drug effects, Tumor Burden immunology, Dendritic Cells immunology, Dendritic Cells pathology, Lung Neoplasms immunology
- Abstract
Checkpoint blockade therapies have improved cancer treatment, but such immunotherapy regimens fail in a large subset of patients. Conventional type 1 dendritic cells (DC1s) control the response to checkpoint blockade in preclinical models and are associated with better overall survival in patients with cancer, reflecting the specialized ability of these cells to prime the responses of CD8
+ T cells1-3 . Paradoxically, however, DC1s can be found in tumours that resist checkpoint blockade, suggesting that the functions of these cells may be altered in some lesions. Here, using single-cell RNA sequencing in human and mouse non-small-cell lung cancers, we identify a cluster of dendritic cells (DCs) that we name 'mature DCs enriched in immunoregulatory molecules' (mregDCs), owing to their coexpression of immunoregulatory genes (Cd274, Pdcd1lg2 and Cd200) and maturation genes (Cd40, Ccr7 and Il12b). We find that the mregDC program is expressed by canonical DC1s and DC2s upon uptake of tumour antigens. We further find that upregulation of the programmed death ligand 1 protein-a key checkpoint molecule-in mregDCs is induced by the receptor tyrosine kinase AXL, while upregulation of interleukin (IL)-12 depends strictly on interferon-γ and is controlled negatively by IL-4 signalling. Blocking IL-4 enhances IL-12 production by tumour-antigen-bearing mregDC1s, expands the pool of tumour-infiltrating effector T cells and reduces tumour burden. We have therefore uncovered a regulatory module associated with tumour-antigen uptake that reduces DC1 functionality in human and mouse cancers.- Published
- 2020
- Full Text
- View/download PDF
39. Macrophage MerTK Promotes Liver Fibrosis in Nonalcoholic Steatohepatitis.
- Author
-
Cai B, Dongiovanni P, Corey KE, Wang X, Shmarakov IO, Zheng Z, Kasikara C, Davra V, Meroni M, Chung RT, Rothlin CV, Schwabe RF, Blaner WS, Birge RB, Valenti L, and Tabas I
- Subjects
- ADAM17 Protein metabolism, Animals, Cell Line, Chronic Disease, Humans, Liver cytology, Macrophages cytology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Rats, Liver metabolism, Liver Cirrhosis metabolism, Macrophages metabolism, Non-alcoholic Fatty Liver Disease metabolism, c-Mer Tyrosine Kinase physiology
- Abstract
Nonalcoholic steatohepatitis (NASH) is emerging as a leading cause of chronic liver disease. However, therapeutic options are limited by incomplete understanding of the mechanisms of NASH fibrosis, which is mediated by activation of hepatic stellate cells (HSCs). In humans, human genetic studies have shown that hypomorphic variations in MERTK, encoding the macrophage c-mer tyrosine kinase (MerTK) receptor, provide protection against liver fibrosis, but the mechanisms remain unknown. We now show that holo- or myeloid-specific Mertk targeting in NASH mice decreases liver fibrosis, congruent with the human genetic data. Furthermore, ADAM metallopeptidase domain 17 (ADAM17)-mediated MerTK cleavage in liver macrophages decreases during steatosis to NASH transition, and mice with a cleavage-resistant MerTK mutant have increased NASH fibrosis. Macrophage MerTK promotes an ERK-TGFβ1 pathway that activates HSCs and induces liver fibrosis. These data provide insights into the role of liver macrophages in NASH fibrosis and provide a plausible mechanism underlying MERTK as a genetic risk factor for NASH fibrosis., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
40. Reinvigorating NIH Grant Peer Review.
- Author
-
Crotty S, Blish C, Cadwell K, Chi H, Goldrath A, Green D, Kaech SM, Krummel M, Pepper M, Rothlin CV, and Wherry EJ
- Subjects
- Humans, National Institutes of Health (U.S.), United States, Peer Review, Research, Research Support as Topic
- Published
- 2020
- Full Text
- View/download PDF
41. Funerals and Feasts: The Immunological Rites of Cell Death.
- Author
-
Galimberti VE, Rothlin CV, and Ghosh S
- Subjects
- Animals, Homeostasis, Humans, Infections pathology, Phagocytosis, Apoptosis immunology
- Abstract
The immune system functions as a vanguard against pathogens and toxins. While it is mostly considered to be activated on the basis of self versus non-self recognition, injury/infection and damage are unavoidably associated with cell death. Does cell death play a role in the regulation of the immune response? Cell death, for better or for worse, is an omnipresent process in all stages of life that are observed throughout most tissues in multicellular organisms. From development to homeostasis in adult organisms, cells commit to scheduled death, while cases of injury and infection result in unscheduled cell death. Novel understanding of the molecular mechanisms that govern cell death demonstrate that, in fact, a plethora of molecular processes participate in directed dying. Parallel to the molecular modalities directing cell death are machineries employed by the organism to respond to dying cells, including either eliciting an inflammatory or immunological response or altogether avoiding it. Disturbing the careful coupling of these two processes is often met with pathology - on one hand a failure to respond to cell death may contribute to the lack of proper immune response or defective development, and on the other hand exaggerated or aberrant response to cell death can trigger unregulated inflammation, autoimmunity, or fibrosis/scarring. Here we review the molecular mechanisms and associated effector responses that accompany some of the most well-known cell death modalities - with an emphasis on efferocytosis, a process by which the dead cell is recognized and engulfed. In doing so, we highlight the TAM (TYRO3, AXL, MERTK) family of receptor tyrosine kinases (RTKs) that functions dually in the recognition and engulfment of dead cells, and as an important negative regulator of inflammation., (Copyright ©2019, Yale Journal of Biology and Medicine.)
- Published
- 2019
42. Bringing on the itch.
- Author
-
Waizman DA, Ghosh S, and Rothlin CV
- Subjects
- Humans, Skin, Dermatitis, Atopic, Pruritus
- Abstract
Neutrophils are the first immune cells that enter the skin and cause itch in atopic dermatitis., Competing Interests: DW, SG, CR No competing interests declared, (© 2019, Waizman et al.)
- Published
- 2019
- Full Text
- View/download PDF
43. aPKC in neuronal differentiation, maturation and function.
- Author
-
Hapak SM, Rothlin CV, and Ghosh S
- Abstract
The atypical Protein Kinase Cs (aPKCs)-PRKCI, PRKCZ and PKMζ-form a subfamily within the Protein Kinase C (PKC) family. These kinases are expressed in the nervous system, including during its development and in adulthood. One of the aPKCs, PKMζ, appears to be restricted to the nervous system. aPKCs are known to play a role in a variety of cellular responses such as proliferation, differentiation, polarity, migration, survival and key metabolic functions such as glucose uptake, that are critical for nervous system development and function. Therefore, these kinases have garnered a lot of interest in terms of their functional role in the nervous system. Here we review the expression and function of aPKCs in neural development and in neuronal maturation and function. Despite seemingly paradoxical findings with genetic deletion versus gene silencing approaches, we posit that aPKCs are likely candidates for regulating many important neurodevelopmental and neuronal functions, and may be associated with a number of human neuropsychiatric diseases., Competing Interests: The authors declare that there are no competing interests associated with the manuscript., (© 2019 The Author(s).)
- Published
- 2019
- Full Text
- View/download PDF
44. Serous macrophages pack Bhlhe40 for a randonnée.
- Author
-
Rothlin CV and Ghosh S
- Subjects
- Cell Proliferation, Homeostasis, Macrophages, Basic Helix-Loop-Helix Transcription Factors, Homeodomain Proteins
- Published
- 2019
- Full Text
- View/download PDF
45. Induction of sterile type 2 inflammation.
- Author
-
de Kouchkovsky DA, Ghosh S, and Rothlin CV
- Subjects
- Agammaglobulinaemia Tyrosine Kinase, Humans, Inflammation, Macrophages, Interleukin-33
- Published
- 2019
- Full Text
- View/download PDF
46. Coagulopathies and inflammatory diseases: '…glimpse of a Snark'.
- Author
-
Del Carmen S, Hapak SM, Ghosh S, and Rothlin CV
- Subjects
- Animals, Humans, Inflammation immunology, Inflammation pathology, Thrombophilia immunology, Thrombophilia pathology, Anticoagulants therapeutic use, Inflammation drug therapy, Pyrazoles therapeutic use, Pyridones therapeutic use, Thrombophilia drug therapy, Warfarin therapeutic use
- Abstract
Coagulopathies and inflammatory diseases, ostensibly, have distinct underlying molecular bases. Notwithstanding, both are host defense mechanisms to physical injury. In invertebrates, clotting can function directly in anti-pathogen defense. Molecules of the vertebrate clotting cascade have also been directly linked to the regulation of inflammation. We posit that thrombophilia may provide resistance against pathogens in vertebrates. The selective pressure of improved anti-pathogen defense may have retained mutations associated with a thrombophilic state in the human population and directly contributed to enhanced inflammation. Indeed, in some inflammatory diseases, at least a subset of patients can be identified as hypercoagulable. Therefore, anticoagulants such as warfarin or apixaban may have a therapeutic role in some inflammatory diseases., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
47. Cenabis Bene: Treg Cells Invite Macrophages to Dine.
- Author
-
Hughes LD, Ghosh S, and Rothlin CV
- Subjects
- Humans, Inflammation, Macrophages immunology, Phagocytosis immunology, T-Lymphocytes, Regulatory immunology
- Abstract
Resolution of the immune response requires a coordinated effort to dampen inflammatory mediators and remove dying cells and debris. In this issue of Immunity, Proto et al. (2018) describe a circuit by which regulatory T cells enhance macrophage consumption of apoptotic cells during resolution., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
48. PAR3-PAR6-atypical PKC polarity complex proteins in neuronal polarization.
- Author
-
Hapak SM, Rothlin CV, and Ghosh S
- Subjects
- Animals, Humans, Models, Neurological, Neurons cytology, Signal Transduction, Adaptor Proteins, Signal Transducing metabolism, Cell Cycle Proteins metabolism, Cell Polarity, Membrane Proteins metabolism, Neurons metabolism, Protein Kinase C metabolism
- Abstract
Polarity is a fundamental feature of cells. Protein complexes, including the PAR3-PAR6-aPKC complex, have conserved roles in establishing polarity across a number of eukaryotic cell types. In neurons, polarity is evident as distinct axonal versus dendritic domains. The PAR3, PAR6, and aPKC proteins also play important roles in neuronal polarization. During this process, either aPKC kinase activity, the assembly of the PAR3-PAR6-aPKC complex or the localization of these proteins is regulated downstream of a number of signaling pathways. In turn, the PAR3, PAR6, and aPKC proteins control various effector molecules to establish neuronal polarity. Herein, we discuss the many signaling mechanisms and effector functions that have been linked to PAR3, PAR6, and aPKC during the establishment of neuronal polarity.
- Published
- 2018
- Full Text
- View/download PDF
49. Axon Regeneration: Antagonistic Signaling Pairs in Neuronal Polarization.
- Author
-
Hapak SM, Ghosh S, and Rothlin CV
- Subjects
- Animals, Central Nervous System physiology, Humans, Recovery of Function physiology, Spinal Cord Injuries physiopathology, Axons physiology, Neurons physiology, Regeneration physiology, Signal Transduction physiology
- Abstract
Genome-wide screens, proteomics, and candidate-based approaches have identified numerous genes associated with neuronal regeneration following central nervous system (CNS) injury. Despite significant progress, functional recovery remains a challenge, even in model systems. Neuronal function depends on segregation of axonal versus dendritic domains. A key to functional recovery may lie in recapitulating the developmental signals that instruct axon specification and growth in adult neurons post-injury. Theoretically, binary activator-inhibitor elements operating as a Turing-like system within neurons can specify axonal versus dendritic domains and promote axon growth. We review here various molecules implicated in axon specification that function as signaling pairs driving neuronal polarization and axon growth., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
50. Antiphospholipid Antibodies Inhibit Trophoblast Toll-Like Receptor and Inflammasome Negative Regulators.
- Author
-
Mulla MJ, Weel IC, Potter JA, Gysler SM, Salmon JE, Peraçoli MTS, Rothlin CV, Chamley LW, and Abrahams VM
- Subjects
- Cell Line, Female, Humans, Immunosuppressive Agents administration & dosage, Inflammation chemically induced, Pregnancy, Pregnancy Trimester, First immunology, Sirolimus administration & dosage, Antibodies, Antiphospholipid immunology, Inflammasomes immunology, NLR Family, Pyrin Domain-Containing 3 Protein immunology, Toll-Like Receptor 4 immunology, Trophoblasts immunology
- Abstract
Objective: Women with antiphospholipid antibodies (aPL) are at risk for pregnancy complications associated with poor placentation and placental inflammation. Although these antibodies are heterogeneous, some anti-β
2 -glycoprotein I (anti-β2 GPI) antibodies can activate Toll-like receptor 4 (TLR-4) and NLRP3 in human first-trimester trophoblasts. The objective of this study was to determine the role of negative regulators of TLR and inflammasome function in aPL-induced trophoblast inflammation., Methods: Human trophoblasts were not treated or were treated with anti-β2 GPI aPL or control IgG in the presence or absence of the common TAM (TYRO3, AXL, and Mer tyrosine kinase [MERTK]) receptor ligand growth arrest-specific protein 6 (GAS6) or the autophagy-inducer rapamycin. The expression and function of the TAM receptor pathway and autophagy were measured by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), Western blotting, and enzyme-linked immunosorbent assay (ELISA). Antiphospholipid antibody-induced trophoblast inflammation was measured by qRT-PCR, activity assays, and ELISA., Results: Anti-β2 GPI aPL inhibited trophoblast TAM receptor function by reducing cellular expression of the receptor tyrosine kinases AXL and MERTK and the ligand GAS6. The addition of GAS6 blocked the effects of aPL on the TLR-4-mediated interleukin-8 (IL-8) response. However, the NLRP3 inflammasome-mediated IL-1β response was not affected by GAS6, suggesting that another regulatory pathway was involved. Indeed, anti-β2 GPI aPL inhibited basal trophoblast autophagy, and reversing this with rapamycin inhibited aPL-induced inflammasome function and IL-1β secretion., Conclusion: Basal TAM receptor function and autophagy may serve to inhibit trophoblast TLR and inflammasome function, respectively. Impairment of TAM receptor signaling and autophagy by anti-β2 GPI aPL may allow subsequent TLR and inflammasome activity, leading to a robust inflammatory response., (© 2018, American College of Rheumatology.)- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.