1
|
de Ligt LA, Gaartman AE, Biemond BJ, Fijnvandraat K, van Bruggen R, Nur E. Neutrophils in sickle cell disease: Exploring their potential role as a therapeutic target. Am J Hematol 2024; 99:1119-1128. [PMID: 38293835 DOI: 10.1002/ajh.27224] [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: 10/11/2023] [Revised: 12/15/2023] [Accepted: 01/01/2024] [Indexed: 02/01/2024]
Abstract
Factors influencing the activation of neutrophils in SCD and the potential neutrophil-mediated ameliorating effects of therapies in SCD.
Collapse
Affiliation(s)
- Lydian A de Ligt
- Amsterdam UMC location University of Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
- Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Department of Pediatric Hematology, Amsterdam, the Netherlands
| | - Aafke E Gaartman
- Amsterdam UMC location University of Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
| | - Bart J Biemond
- Amsterdam UMC location University of Amsterdam, Department of Hematology, Amsterdam, The Netherlands
| | - Karin Fijnvandraat
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
- Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Department of Pediatric Hematology, Amsterdam, the Netherlands
| | - Robin van Bruggen
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
| | - Erfan Nur
- Amsterdam UMC location University of Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Jang YS, Park SH, Kang SG, Lee JS, Ko HJ, Kim PH. Combined Treatment With TGF-β1, Retinoic Acid, and Lactoferrin Robustly Generate Inducible Tregs (iTregs) Against High Affinity Ligand. Immune Netw 2023; 23:e37. [PMID: 37970231 PMCID: PMC10643331 DOI: 10.4110/in.2023.23.e37] [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: 03/20/2023] [Revised: 08/25/2023] [Accepted: 09/06/2023] [Indexed: 11/17/2023] Open
Abstract
Forkhead box P3-positive (Foxp3+)-inducible Tregs (iTregs) are readily generated by TGF-β1 at low TCR signaling intensity. TGF-β1-mediated Foxp3 expression is further enhanced by retinoic acid (RA) and lactoferrin (LF). However, the intensity of TCR signaling required for induction of Foxp3 expression by TGF-β1 in combination with RA and LF is unknown. Here, we found that either RA or LF alone decreased TGF-β1-mediated Foxp3 expression at low TCR signaling intensity. In contrast, at high TCR signaling intensity, the addition of either RA or LF strongly increased TGF-β1-mediated Foxp3 expression. Moreover, decreased CD28 stimulation was more favorable for TGF-β1/LF-mediated Foxp3 expression. Lastly, we found that at high signaling intensities of both TCR and CD28, combined treatment with TGF-β1, RA, and LF induced robust expression of Foxp3, in parallel with powerful suppressive activity against responder T cell proliferation. Our findings that TGFβ/RA/LF strongly generate high affinity Ag-specific iTreg population would be useful for the control of unwanted hypersensitive immune reactions such as various autoimmune diseases.
Collapse
Affiliation(s)
- Young-Saeng Jang
- Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Korea
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 24341, Korea
| | - Sun-Hee Park
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 24341, Korea
| | - Seung-Goo Kang
- Division of Biomedical Convergence, School of Biomedical Science, Kangwon National University, Chuncheon 24341, Korea
| | - Jung-Shin Lee
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 24341, Korea
| | - Hyun-Jeong Ko
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Pyeung-Hyeun Kim
- Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Korea
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
3
|
Jiang H, Shin DH, Yi Y, Fan X, Gumin J, He J, Gillard AG, Lang FF, Gomez-Manzano C, Fueyo J. Adjuvant Therapy with Oncolytic Adenovirus Delta-24-RGDOX After Intratumoral Adoptive T-cell Therapy Promotes Antigen Spread to Sustain Systemic Antitumor Immunity. CANCER RESEARCH COMMUNICATIONS 2023; 3:1118-1131. [PMID: 37379361 PMCID: PMC10295804 DOI: 10.1158/2767-9764.crc-23-0054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 06/30/2023]
Abstract
Cancer cell heterogeneity and immunosuppressive tumor microenvironment (TME) pose a challenge in treating solid tumors with adoptive cell therapies targeting limited tumor-associated antigens (TAA), such as chimeric antigen receptor T-cell therapy. We hypothesize that oncolytic adenovirus Delta-24-RGDOX activates the TME and promote antigen spread to potentiate the abscopal effect of adoptive TAA-targeting T cells in localized intratumoral treatment. Herein, we used C57BL/6 mouse models with disseminated tumors derived from B16 melanoma cell lines to assess therapeutic effects and antitumor immunity. gp100-specific pmel-1 or ovalbumin (OVA)-specific OT-I T cells were injected into the first subcutaneous tumor, followed by three injections of Delta-24-RGDOX. We found TAA-targeting T cells injected into one subcutaneous tumor showed tumor tropism. Delta-24-RGDOX sustained the systemic tumor regression mediated by the T cells, leading to improved survival rate. Further analysis revealed that, in mice with disseminated B16-OVA tumors, Delta-24-RGDOX increased CD8+ leukocyte density within treated and untreated tumors. Importantly, Delta-24-RGDOX significantly reduced the immunosuppression of endogenous OVA-specific CTLs while increasing that of CD8+ leukocytes and, to a lesser extent, adoptive pmel-1 T cells. Consequently, Delta-24-RGDOX drastically increased the density of the OVA-specific CTLs in both tumors, and the combination synergistically enhanced the effect. Consistently, the splenocytes from the combination group showed a significantly stronger response against other TAAs (OVA and TRP2) than gp100, resulted in higher activity against tumor cells. Therefore, our data demonstrate that, as an adjuvant therapy followed TAA-targeting T cells in localized treatment, Delta-24-RGDOX activates TME and promotes antigen spread, leading to efficacious systemic antitumor immunity to overcome tumor relapse. Significance Adjuvant therapy with oncolytic viruses promotes antigen spread to potentiate localized intratumoral adoptive T-cell therapy with limited TAA targets, leading to sustainable systemic antitumor immunity to overcome tumor relapse.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dong Ho Shin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yanhua Yi
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xuejun Fan
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joy Gumin
- Department of Neuro-Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiasen He
- Pediatric division, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Andrew G. Gillard
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frederick F. Lang
- Department of Neuro-Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
4
|
Kokkinou E, Pandey RV, Mazzurana L, Gutierrez-Perez I, Tibbitt CA, Weigel W, Soini T, Carrasco A, Rao A, Nagasawa M, Bal SM, Jangard M, Friberg D, Lindforss U, Nordenvall C, Ljunggren M, Haapaniemi S, Keita ÅV, Söderholm J, Hedin C, Spits H, Bryceson YT, Mjösberg J. CD45RA +CD62L - ILCs in human tissues represent a quiescent local reservoir for the generation of differentiated ILCs. Sci Immunol 2022; 7:eabj8301. [PMID: 35427178 DOI: 10.1126/sciimmunol.abj8301] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Innate lymphoid cells (ILCs) are highly plastic and predominantly mucosal tissue-resident cells that contribute to both homeostasis and inflammation depending on the microenvironment. The discovery of naïve-like ILCs suggests an ILC differentiation process that is akin to naïve T cell differentiation. Delineating the mechanisms that underlie ILC differentiation in tissues is crucial for understanding ILC biology in health and disease. Here, we showed that tonsillar ILCs expressing CD45RA lacked proliferative activity, indicative of cellular quiescence. CD62L distinguished two subsets of CD45RA+ ILCs. CD45RA+CD62L+ ILCs (CD62L+ ILCs) resembled circulating naïve ILCs because they lacked the transcriptional, metabolic, epigenetic, and cytokine production signatures of differentiated ILCs. CD45RA+CD62L- ILCs (CD62L- ILCs) were epigenetically similar to CD62L+ ILCs but showed a transcriptional, metabolic, and cytokine production signature that was more akin to differentiated ILCs. CD62L+ and CD62L- ILCs contained uni- and multipotent precursors of ILC1s/NK cells and ILC3s. Differentiation of CD62L+ and CD62L- ILCs led to metabolic reprogramming including up-regulation of genes associated with glycolysis, which was needed for their effector functions after differentiation. CD62L- ILCs with preferential differentiation capacity toward IL-22-producing ILC3s accumulated in the inflamed mucosa of patients with inflammatory bowel disease. These data suggested distinct differentiation potential of CD62L+ and CD62L- ILCs between tissue microenvironments and identified that manipulation of these cells is a possible approach to restore tissue-immune homeostasis.
Collapse
Affiliation(s)
- Efthymia Kokkinou
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Ram Vinay Pandey
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Luca Mazzurana
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Irene Gutierrez-Perez
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Christopher Andrew Tibbitt
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Whitney Weigel
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Tea Soini
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Anna Carrasco
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Anna Rao
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Maho Nagasawa
- Department of Experimental Immunology, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Suzanne M Bal
- Department of Experimental Immunology, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Mattias Jangard
- ENT Unit, Sophiahemmet University Research Laboratory and Sophiahemmet Hospital, Stockholm, Sweden
| | - Danielle Friberg
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Ulrik Lindforss
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Department of Pelvic Cancer, GI Oncology and Colorectal Surgery Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Nordenvall
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Department of Pelvic Cancer, GI Oncology and Colorectal Surgery Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Malin Ljunggren
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Department of Pelvic Cancer, GI Oncology and Colorectal Surgery Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Staffan Haapaniemi
- Department of Surgery, Vrinnevi Hospital, Norrköping, Sweden.,Department of Surgery, Linköping University, Linköping, Sweden
| | - Åsa V Keita
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johan Söderholm
- Department of Surgery, Linköping University, Linköping, Sweden.,Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Charlotte Hedin
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Gastroenterology Unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Hergen Spits
- Department of Experimental Immunology, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Yenan T Bryceson
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Jenny Mjösberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
5
|
Kajikawa M, Imaizumi N, Machii S, Nakamura T, Harigane N, Kimura M, Miyano K, Ishido S, Kanamoto T. Kaposi's sarcoma-associated herpesvirus ubiquitin ligases downregulate cell surface expression of l-selectin. J Gen Virol 2021; 102. [PMID: 34726593 DOI: 10.1099/jgv.0.001678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic etiological factor for Kaposi's sarcoma and primary effusion lymphoma in immunocompromised patients. KSHV utilizes two immune evasion E3 ubiquitin ligases, namely K3 and K5, to downregulate the expression of antigen-presenting molecules and ligands of natural killer (NK) cells in the host cells through an ubiquitin-dependent endocytic mechanism. This allows the infected cells to evade surveillance and elimination by cytotoxic lymphocytes and NK cells. The number of host cell molecular substrates reported for these ubiquitin ligases is limited. The identification of novel substrates for these ligases will aid in elucidating the mechanism underlying immune evasion of KSHV. This study demonstrated that K5 downregulated the cell surface expression of l-selectin, a C-type lectin-like adhesion receptor expressed in the lymphocytes. Tryptophan residue located at the centre of the E2-binding site in the K5 RINGv domain was essential to downregulate l-selectin expression. Additionally, the lysine residues located at the cytoplasmic tail of l-selectin were required for the K5-mediated downregulation of l-selectin. K5 promoted the degradation of l-selectin through polyubiquitination. These results suggest that K5 downregulates l-selectin expression on the cell surface by promoting polyubiquitination and ubiquitin-dependent endocytosis, which indicated that l-selectin is a novel substrate for K5. Additionally, K3 downregulated l-selectin expression. The findings of this study will aid in the elucidation of a novel immune evasion mechanism in KSHV.
Collapse
Affiliation(s)
- Mizuho Kajikawa
- Laboratory of Microbiology, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan
| | - Nanae Imaizumi
- Laboratory of Microbiology, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan
| | - Shiho Machii
- Laboratory of Microbiology, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan
| | - Tomoka Nakamura
- Laboratory of Microbiology, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan
| | - Nana Harigane
- Laboratory of Microbiology, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan
| | - Minako Kimura
- Laboratory of Microbiology, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan
| | - Kei Miyano
- Department of Biochemistry, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| | - Satoshi Ishido
- Department of Microbiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Taisei Kanamoto
- Laboratory of Microbiology, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan
| |
Collapse
|
6
|
Rahman I, Collado Sánchez A, Davies J, Rzeniewicz K, Abukscem S, Joachim J, Hoskins Green HL, Killock D, Sanz MJ, Charras G, Parsons M, Ivetic A. L-selectin regulates human neutrophil transendothelial migration. J Cell Sci 2021; 134:jcs.250340. [PMID: 33408247 PMCID: PMC7888707 DOI: 10.1242/jcs.250340] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 12/03/2020] [Indexed: 01/13/2023] Open
Abstract
The migration of circulating neutrophils towards damaged or infected tissue is absolutely critical to the inflammatory response. L-selectin is a cell adhesion molecule abundantly expressed on circulating neutrophils. For over two decades, neutrophil L-selectin has been assigned the exclusive role of supporting tethering and rolling – the initial stages of the multi-step adhesion cascade. Here, we provide direct evidence for L-selectin contributing to neutrophil transendothelial migration (TEM). We show that L-selectin co-clusters with PECAM-1 – a well-characterised cell adhesion molecule involved in regulating neutrophil TEM. This co-clustering behaviour occurs specifically during TEM, which serves to augment ectodomain shedding of L-selectin and expedite the time taken for TEM (TTT) to complete. Blocking PECAM-1 signalling (through mutation of its cytoplasmic tail), PECAM-1-dependent adhesion or L-selectin shedding, leads to a significant delay in the TTT. Finally, we show that co-clustering of L-selectin with PECAM-1 occurs specifically across TNF- but not IL-1β-activated endothelial monolayers – implying unique adhesion interactomes forming in a cytokine-specific manner. To our knowledge, this is the first report to implicate a non-canonical role for L-selectin in regulating neutrophil TEM. Highlighted Article: Neutrophil L-selectin co-clusters with PECAM-1 in cis during transendothelial migration (TEM). Clustering neutrophil PECAM-1 activates p38 MAPK and JNK to regulate L-selectin shedding, which in turn expedites TEM.
Collapse
Affiliation(s)
- Izajur Rahman
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Aida Collado Sánchez
- Department of Pharmacology and Faculty of Medicine and Odontology, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain.,Institute of Health Research INCLIVA, University Clinic Hospital of Valencia, Av. Menéndez Pelayo 4, 46010, Valencia, Spain
| | - Jessica Davies
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Karolina Rzeniewicz
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Sarah Abukscem
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Justin Joachim
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Hannah L Hoskins Green
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - David Killock
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Maria Jesus Sanz
- Department of Pharmacology and Faculty of Medicine and Odontology, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain.,Institute of Health Research INCLIVA, University Clinic Hospital of Valencia, Av. Menéndez Pelayo 4, 46010, Valencia, Spain.,CIBERDEM-Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, ISCIII, Av. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Guillaume Charras
- London Centre for Nanotechnology, University College London, London WC1H 0AH, UK
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Aleksandar Ivetic
- BHF Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
7
|
Prakash S, Roy S, Srivastava R, Coulon PG, Dhanushkodi NR, Vahed H, Jankeel A, Geertsema R, Amezquita C, Nguyen L, Messaoudi I, Burkhardt AM, BenMohamed L. Unique molecular signatures of antiviral memory CD8 + T cells associated with asymptomatic recurrent ocular herpes. Sci Rep 2020; 10:13843. [PMID: 32796943 PMCID: PMC7427992 DOI: 10.1038/s41598-020-70673-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
The nature of antiviral CD8+ T cells associated with protective and pathogenic herpes simplex virus type 1 (HSV-1) infections remains unclear. We compared the transcriptome, phenotype, and function of memory CD8+ T cells, sharing the same HSV-1 epitope-specificities, from infected HLA-A*0201 positive symptomatic (SYMP) vs. asymptomatic (ASYMP) individuals and HLA-A*0201 transgenic rabbits. Compared to higher frequencies of multifunctional effector memory CD8+ TEM cells in ASYMP individuals, the SYMP individuals presented dysfunctional CD8+ TEM cells, expressing major exhaustion pathways. Compared to protected ASYMP HLA transgenic rabbits, the trigeminal ganglia of non-protected SYMP HLA transgenic rabbits had higher frequencies of dysfunctional tissue-resident CD8+ TRM cells. Moreover, blockade of T cell exhaustion pathways restored the function of CD8+ T cells, reduced virus reactivation, and diminished recurrent disease in HLA transgenic rabbits. These findings reveal unique molecular signatures of protective CD8+ T cells and pave the way for T-cell-based immunotherapy to combat recurrent ocular herpes.
Collapse
Affiliation(s)
- Swayam Prakash
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Soumyabrata Roy
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Ruchi Srivastava
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Pierre-Gregoire Coulon
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Nisha R Dhanushkodi
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Hawa Vahed
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Allen Jankeel
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA, 92697, USA
| | - Roger Geertsema
- University Laboratory Animal Resources, University of California Irvine, Irvine, CA, 92697, USA
| | - Cassandra Amezquita
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Lan Nguyen
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA, 92697, USA
| | - Amanda M Burkhardt
- Vaccine Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, CA, 92617, USA
- Institute for Immunology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - Lbachir BenMohamed
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA.
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA, 92697, USA.
- Vaccine Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, CA, 92617, USA.
- Institute for Immunology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
8
|
Newe A, Rzeniewicz K, König M, Schroer CFE, Joachim J, Rey-Gallardo A, Marrink SJ, Deka J, Parsons M, Ivetic A. Serine Phosphorylation of L-Selectin Regulates ERM Binding, Clustering, and Monocyte Protrusion in Transendothelial Migration. Front Immunol 2019; 10:2227. [PMID: 31608057 PMCID: PMC6774396 DOI: 10.3389/fimmu.2019.02227] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 09/03/2019] [Indexed: 12/15/2022] Open
Abstract
The migration of circulating leukocytes toward damaged tissue is absolutely fundamental to the inflammatory response, and transendothelial migration (TEM) describes the first cellular barrier that is breached in this process. Human CD14+ inflammatory monocytes express L-selectin, bestowing a non-canonical role in invasion during TEM. In vivo evidence supports a role for L-selectin in regulating TEM and chemotaxis, but the intracellular mechanism is poorly understood. The ezrin-radixin-moesin (ERM) proteins anchor transmembrane proteins to the cortical actin-based cytoskeleton and additionally act as signaling adaptors. During TEM, the L-selectin tail within transmigrating pseudopods interacts first with ezrin to transduce signals for protrusion, followed by moesin to drive ectodomain shedding of L-selectin to limit protrusion. Collectively, interaction of L-selectin with ezrin and moesin fine-tunes monocyte protrusive behavior in TEM. Using FLIM/FRET approaches, we show that ERM binding is absolutely required for outside-in L-selectin clustering. The cytoplasmic tail of human L-selectin contains two serine (S) residues at positions 364 and 367, and here we show that they play divergent roles in regulating ERM binding. Phospho-S364 blocks direct interaction with ERM, whereas molecular modeling suggests phospho-S367 likely drives desorption of the L-selectin tail from the inner leaflet of the plasma membrane to potentiate ERM binding. Serine-to-alanine mutagenesis of S367, but not S364, significantly reduced monocyte protrusive behavior in TEM under flow conditions. Our data propose a model whereby L-selectin tail desorption from the inner leaflet of the plasma membrane and ERM binding are two separable steps that collectively regulate protrusive behavior in TEM.
Collapse
Affiliation(s)
- Abigail Newe
- BHF Centre of Research Excellence, James Black Centre, King's College London, London, United Kingdom
| | - Karolina Rzeniewicz
- BHF Centre of Research Excellence, James Black Centre, King's College London, London, United Kingdom
| | - Melanie König
- Groningen Biomolecular Sciences and Biotechnology Institute, Groningen, Netherlands
| | - Carsten F E Schroer
- Groningen Biomolecular Sciences and Biotechnology Institute, Groningen, Netherlands
| | - Justin Joachim
- BHF Centre of Research Excellence, James Black Centre, King's College London, London, United Kingdom
| | - Angela Rey-Gallardo
- BHF Centre of Research Excellence, James Black Centre, King's College London, London, United Kingdom
| | - Siewert J Marrink
- Groningen Biomolecular Sciences and Biotechnology Institute, Groningen, Netherlands
| | - Jürgen Deka
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Aleksandar Ivetic
- BHF Centre of Research Excellence, James Black Centre, King's College London, London, United Kingdom
| |
Collapse
|
9
|
Ivetic A, Hoskins Green HL, Hart SJ. L-selectin: A Major Regulator of Leukocyte Adhesion, Migration and Signaling. Front Immunol 2019; 10:1068. [PMID: 31139190 PMCID: PMC6527602 DOI: 10.3389/fimmu.2019.01068] [Citation(s) in RCA: 299] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/26/2019] [Indexed: 12/12/2022] Open
Abstract
L-selectin (CD62L) is a type-I transmembrane glycoprotein and cell adhesion molecule that is expressed on most circulating leukocytes. Since its identification in 1983, L-selectin has been extensively characterized as a tethering/rolling receptor. There is now mounting evidence in the literature to suggest that L-selectin plays a role in regulating monocyte protrusion during transendothelial migration (TEM). The N-terminal calcium-dependent (C-type) lectin domain of L-selectin interacts with numerous glycans, including sialyl Lewis X (sLex) for tethering/rolling and proteoglycans for TEM. Although the signals downstream of L-selectin-dependent adhesion are poorly understood, they will invariably involve the short 17 amino acid cytoplasmic tail. In this review we will detail the expression of L-selectin in different immune cell subsets, and its influence on cell behavior. We will list some of the diverse glycans known to support L-selectin-dependent adhesion, within luminal and abluminal regions of the vessel wall. We will describe how each domain within L-selectin contributes to adhesion, migration and signal transduction. A significant focus on the L-selectin cytoplasmic tail and its proposed contribution to signaling via the ezrin-radixin-moesin (ERM) family of proteins will be outlined. Finally, we will discuss how ectodomain shedding of L-selectin during monocyte TEM is essential for the establishment of front-back cell polarity, bestowing emigrated cells the capacity to chemotax toward sites of damage.
Collapse
Affiliation(s)
- Aleksandar Ivetic
- King's College London, School of Cardiovascular Medicine and Sciences, BHF Center of Research Excellence, London, United Kingdom
| | - Hannah Louise Hoskins Green
- King's College London, School of Cardiovascular Medicine and Sciences, BHF Center of Research Excellence, London, United Kingdom
| | - Samuel James Hart
- King's College London, School of Cardiovascular Medicine and Sciences, BHF Center of Research Excellence, London, United Kingdom
| |
Collapse
|
10
|
Mohammed RN, Wehenkel SC, Galkina EV, Yates EK, Preece G, Newman A, Watson HA, Ohme J, Bridgeman JS, Durairaj RRP, Moon OR, Ladell K, Miners KL, Dolton G, Troeberg L, Kashiwagi M, Murphy G, Nagase H, Price DA, Matthews RJ, Knäuper V, Ager A. ADAM17-dependent proteolysis of L-selectin promotes early clonal expansion of cytotoxic T cells. Sci Rep 2019; 9:5487. [PMID: 30940840 PMCID: PMC6445073 DOI: 10.1038/s41598-019-41811-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 03/12/2019] [Indexed: 01/15/2023] Open
Abstract
L-selectin on T-cells is best known as an adhesion molecule that supports recruitment of blood-borne naïve and central memory cells into lymph nodes. Proteolytic shedding of the ectodomain is thought to redirect activated T-cells from lymph nodes to sites of infection. However, we have shown that activated T-cells re-express L-selectin before lymph node egress and use L-selectin to locate to virus-infected tissues. Therefore, we considered other roles for L-selectin proteolysis during T cell activation. In this study, we used T cells expressing cleavable or non-cleavable L-selectin and determined the impact of L-selectin proteolysis on T cell activation in virus-infected mice. We confirm an essential and non-redundant role for ADAM17 in TCR-induced proteolysis of L-selectin in mouse and human T cells and show that L-selectin cleavage does not regulate T cell activation measured by CD69 or TCR internalisation. Following virus infection of mice, L-selectin proteolysis promoted early clonal expansion of cytotoxic T cells resulting in an 8-fold increase over T cells unable to cleave L-selectin. T cells unable to cleave L-selectin showed delayed proliferation in vitro which correlated with lower CD25 expression. Based on these results, we propose that ADAM17-dependent proteolysis of L-selectin should be considered a regulator of T-cell activation at sites of immune activity.
Collapse
Affiliation(s)
- Rebar N Mohammed
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
- College of Veterinary Medicine, University of Sulaimani, Sulaimani, Kurdistan, Iraq
| | - Sophie C Wehenkel
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Elena V Galkina
- Francis Crick Institute, London, NW1 1AT, UK
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | | | | | - Andrew Newman
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - H Angharad Watson
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Julia Ohme
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - John S Bridgeman
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Ruban R P Durairaj
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Owen R Moon
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Kristin Ladell
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Kelly L Miners
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Garry Dolton
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Linda Troeberg
- Norwich Medical School, University of East Anglia, Norwich, NR4 7UQ, UK
| | | | - Gillian Murphy
- University of Cambridge Depratment of Oncology, Cancer Research UK Cambridge Insitute, Li Ka Shing Centre, Cambridge, CB2 0RE, UK
| | - Hideaki Nagase
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - David A Price
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - R James Matthews
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Vera Knäuper
- School of Dentistry, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Ann Ager
- Divsion of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK.
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK.
| |
Collapse
|
11
|
Nawaz F, Goes LR, Ray JC, Olowojesiku R, Sajani A, Ansari AA, Perrone I, Hiatt J, Van Ryk D, Wei D, Waliszewski M, Soares MA, Jelicic K, Connors M, Migueles SA, Martinelli E, Villinger F, Cicala C, Fauci AS, Arthos J. MAdCAM costimulation through Integrin-α 4β 7 promotes HIV replication. Mucosal Immunol 2018; 11:1342-1351. [PMID: 29875402 PMCID: PMC6160318 DOI: 10.1038/s41385-018-0044-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 03/16/2018] [Accepted: 04/04/2018] [Indexed: 02/04/2023]
Abstract
Human gut-associated lymphoid tissues (GALT) play a key role in the acute phase of HIV infection. The propensity of HIV to replicate in these tissues, however, is not fully understood. Access and migration of naive and memory CD4+ T cells to these sites is mediated by interactions between integrin α4β7, expressed on CD4+ T cells, and MAdCAM, expressed on high endothelial venules. We report here that MAdCAM delivers a potent costimulatory signal to naive and memory CD4+ T cells following ligation with α4β7. Such costimulation promotes high levels of HIV replication. An anti-α4β7 mAb that prevents mucosal transmission of SIV blocks MAdCAM signaling through α4β7 and MAdCAM-dependent viral replication. MAdCAM costimulation of memory CD4+ T cells is sufficient to drive cellular proliferation and the upregulation of CCR5, while naive CD4+ T cells require both MAdCAM and retinoic acid to achieve the same response. The pairing of MAdCAM and retinoic acid is unique to the GALT, leading us to propose that HIV replication in these sites is facilitated by MAdCAM-α4β7 interactions. Moreover, complete inhibition of MAdCAM signaling by an anti-α4β7 mAb, an analog of the clinically approved therapeutic vedolizumab, highlights the potential of such agents to control acute HIV infection.
Collapse
Affiliation(s)
- Fatima Nawaz
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Livia R Goes
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
- Programa de Genética, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Jocelyn C Ray
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Ronke Olowojesiku
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Alia Sajani
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Aftab A Ansari
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ian Perrone
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Joseph Hiatt
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Donald Van Ryk
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Danlan Wei
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Mia Waliszewski
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Marcelo A Soares
- Programa de Genética, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Departamento de Genética, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Katija Jelicic
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Mark Connors
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Stephen A Migueles
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Elena Martinelli
- Center of Biomedical Research, Population Council, New York, NY, 10017, USA
| | - Francois Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, 70560, USA
| | - Claudia Cicala
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Anthony S Fauci
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - James Arthos
- Laboratory of Immunoregulation, National Institutes of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA.
| |
Collapse
|
12
|
Ivetic A. A head-to-tail view of L-selectin and its impact on neutrophil behaviour. Cell Tissue Res 2018; 371:437-453. [PMID: 29353325 PMCID: PMC5820395 DOI: 10.1007/s00441-017-2774-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/05/2017] [Indexed: 01/04/2023]
Abstract
L-selectin is a type I transmembrane cell adhesion molecule expressed on most circulating leukocytes, including neutrophils. Engagement of L-selectin with endothelial-derived ligands initiates neutrophil tethering and rolling behaviour along luminal walls of post-capillary venules, constituting the first step of the multi-step adhesion cascade. There is a large body of evidence to suggest that signalling downstream of L-selectin can influence neutrophil behaviour: adhesion, migration and priming. This review will cover aspects of L-selectin form and function and introduce the “triad of L-selectin regulation”, highlighting the inextricable links between adhesion, signalling and ectodomain shedding and also highlighting the cytosolic proteins that interconnect them. Recent advances in how L-selectin impacts priming, transendothelial migration (TEM) and cell polarity will also be discussed.
Collapse
Affiliation(s)
- Aleksandar Ivetic
- BHF Centre for Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, James Black Centre 125, Coldharbour Lane, London, SE5 9NU, UK.
| |
Collapse
|
13
|
SDA and IDA - Two aptamers to inhibit cancer cell adhesion. Biochimie 2017; 145:84-90. [PMID: 29080832 DOI: 10.1016/j.biochi.2017.10.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 10/24/2017] [Indexed: 02/07/2023]
Abstract
Aptamers which bind to proteins involved in cell-cell interactions could have significant value to directly affect cancer cell adhesion or for directed cargo delivery. Here, I discuss two aptamers: aptamer SDA which binds to E- and P-selectin, and aptamer IDA which binds to α6β4 integrin. Both aptamers (SDA 91 nt and IDA 77 nt) bind their target proteins with dissociation constants in the 100-150 nM range and substantially inhibit special cellular adhesion, possibly a first and pivotal step in transendothelial migration during metastasis formation. The aptamers' half-lives in cell culture media are between two and six hours. IDA is internalized by integrin presenting cells within minutes thus possibly serving as vehicle for directed cargo delivery.
Collapse
|
14
|
Hobbs SJ, Nolz JC. Regulation of T Cell Trafficking by Enzymatic Synthesis of O-Glycans. Front Immunol 2017; 8:600. [PMID: 28596771 PMCID: PMC5442166 DOI: 10.3389/fimmu.2017.00600] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/08/2017] [Indexed: 12/27/2022] Open
Abstract
Selectins constitute a family of oligosaccharide binding proteins that play critical roles in regulating the trafficking of leukocytes. In T cells, L-selectin (CD62L) controls the capacity for naive and memory T cells to actively survey peripheral lymph nodes, whereas P- and E-selectin capture activated T cells on inflamed vascular endothelium to initiate extravasation into non-lymphoid tissues. The capacity for T cells to interact with all of these selectins is dependent on the enzymatic synthesis of complex O-glycans, and thus, this protein modification plays an indispensable role in regulating the distribution and homing of both naive and previously activated T cells in vivo. In contrast to neutrophils, O-glycan synthesis is highly dynamic in T cell populations and is largely controlled by extracellular stimuli such as antigen recognition or signaling though cytokine receptors. Herein, we review the basic principles of enzymatic synthesis of complex O-glycans, discuss tools and reagents for studying this type of protein modification and highlight our current understanding of how O-glycan synthesis is regulated and subsequently impacts the trafficking potential of diverse T cell populations.
Collapse
Affiliation(s)
- Samuel J Hobbs
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, United States
| | - Jeffrey C Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, United States.,Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, United States.,Department of Radiation Medicine, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
15
|
Margaryan S, Hyusyan A, Martirosyan A, Sargsian S, Manukyan G. Differential modulation of innate immune response by epinephrine and estradiol. Horm Mol Biol Clin Investig 2017; 30:/j/hmbci.ahead-of-print/hmbci-2016-0046/hmbci-2016-0046.xml. [PMID: 28475489 DOI: 10.1515/hmbci-2016-0046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/29/2016] [Indexed: 11/15/2022]
Abstract
Background Although it is widely accepted that catecholamines and estrogens influence immunity and have consequences for health, their effect on innate immunity (e.g. monocytes and neutrophils) is still not fully investigated. Materials and methods Our study aimed to analyze the production of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, monocyte chemoattractant protein (MCP)-1 and IL-8 by whole blood cells following short-term exposure to epinephrine (Epi) and 17β-estradiol (E2) in the presence or absence of lipopolysaccharide (LPS). We also evaluated the in vitro effect of these hormones on expression of β2 integrin (CD11b/CD18) and L-selectin (CD62L) by circulating neutrophils and monocytes in the blood of healthy subjects. Results Epi has shown a potential to modulate the production of pro-inflammatory mediators. Its exposure resulted in significantly increased production of IL-8 in a dose-dependent manner. On the contrary, a dose-dependent suppression of LPS-induced production of IL-1β, IL-8, and MCP-1 by Epi was observed. In neutrophils, a modest rise in CD11b expression was observed after Epi exposure. Simultaneously, Epi suppressed LPS-induced expression of CD11b and CD18. In monocytes, Epi suppressed LPS-induced expression of C11b. E2 inhibited LPS-induced TNF-α production and caused a significant decrease in CD62L expression in both cell populations. No significant changes were observed after double exposure of cells with Epi and E2. Conclusions Thus, our results show that Epi and E2 differentially modulate the innate immune response and have a dual effect on cytokine modulation. The findings suggest that the observed immunoregulatory role of Epi and E2 may influence the outcome in endotoxin responses and can be critical in the regulation of inflammatory responses.
Collapse
|
16
|
Francis A, Bosio E, Stone SF, Fatovich DM, Arendts G, Nagree Y, Macdonald SPJ, Mitenko H, Rajee M, Burrows S, Brown SGA. Neutrophil activation during acute human anaphylaxis: analysis of MPO and sCD62L. Clin Exp Allergy 2017; 47:361-370. [DOI: 10.1111/cea.12868] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/14/2016] [Accepted: 11/18/2016] [Indexed: 12/01/2022]
Affiliation(s)
- A. Francis
- Centre for Clinical Research in Emergency Medicine; Harry Perkins Institute of Medical Research; Perth WA Australia
- Discipline of Emergency Medicine; School of Primary; Aboriginal and Rural Health Care; University of Western Australia; Crawley WA Australia
| | - E. Bosio
- Centre for Clinical Research in Emergency Medicine; Harry Perkins Institute of Medical Research; Perth WA Australia
- Discipline of Emergency Medicine; School of Primary; Aboriginal and Rural Health Care; University of Western Australia; Crawley WA Australia
| | - S. F. Stone
- Centre for Clinical Research in Emergency Medicine; Harry Perkins Institute of Medical Research; Perth WA Australia
- Discipline of Emergency Medicine; School of Primary; Aboriginal and Rural Health Care; University of Western Australia; Crawley WA Australia
| | - D. M. Fatovich
- Centre for Clinical Research in Emergency Medicine; Harry Perkins Institute of Medical Research; Perth WA Australia
- Discipline of Emergency Medicine; School of Primary; Aboriginal and Rural Health Care; University of Western Australia; Crawley WA Australia
- Emergency Department; Royal Perth Hospital; Perth WA Australia
| | - G. Arendts
- Centre for Clinical Research in Emergency Medicine; Harry Perkins Institute of Medical Research; Perth WA Australia
- Discipline of Emergency Medicine; School of Primary; Aboriginal and Rural Health Care; University of Western Australia; Crawley WA Australia
- Emergency Department; Royal Perth Hospital; Perth WA Australia
- Emergency Department, Fiona Stanley Hospital; Murdoch WA Australia
| | - Y. Nagree
- Centre for Clinical Research in Emergency Medicine; Harry Perkins Institute of Medical Research; Perth WA Australia
- Emergency Department, Fiona Stanley Hospital; Murdoch WA Australia
- Emergency Department; Fremantle Hospital; Fremantle WA Australia
| | - S. P. J. Macdonald
- Centre for Clinical Research in Emergency Medicine; Harry Perkins Institute of Medical Research; Perth WA Australia
- Discipline of Emergency Medicine; School of Primary; Aboriginal and Rural Health Care; University of Western Australia; Crawley WA Australia
- Emergency Department; Royal Perth Hospital; Perth WA Australia
- Emergency Department; Armadale Kelmscott Memorial Hospital; Mount Nasura WA Australia
| | - H. Mitenko
- Centre for Clinical Research in Emergency Medicine; Harry Perkins Institute of Medical Research; Perth WA Australia
- Emergency Department; South West Health Campus; Bunbury WA Australia
| | - M. Rajee
- Centre for Clinical Research in Emergency Medicine; Harry Perkins Institute of Medical Research; Perth WA Australia
- Emergency Department; Austin Hospital; Heidelberg VIC Australia
| | - S. Burrows
- School of Medicine & Pharmacology; University of Western Australia; Perth WA Australia
| | - S. G. A. Brown
- Centre for Clinical Research in Emergency Medicine; Harry Perkins Institute of Medical Research; Perth WA Australia
- Discipline of Emergency Medicine; School of Primary; Aboriginal and Rural Health Care; University of Western Australia; Crawley WA Australia
- Emergency Department; Royal Perth Hospital; Perth WA Australia
- Emergency Department; Royal Hobart Hospital; Hobart TAS Australia
| |
Collapse
|
17
|
Braley A, Kwak T, Jules J, Harja E, Landgraf R, Hudson BI. Regulation of Receptor for Advanced Glycation End Products (RAGE) Ectodomain Shedding and Its Role in Cell Function. J Biol Chem 2016; 291:12057-73. [PMID: 27022018 DOI: 10.1074/jbc.m115.702399] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Indexed: 01/11/2023] Open
Abstract
The receptor for advanced glycation end products (RAGE) is a multiligand transmembrane receptor that can undergo proteolysis at the cell surface to release a soluble ectodomain. Here we observed that ectodomain shedding of RAGE is critical for its role in regulating signaling and cellular function. Ectodomain shedding of both human and mouse RAGE was dependent on ADAM10 activity and induced with chemical activators of shedding (ionomycin, phorbol 12-myristate 13-acetate, and 4-aminophenylmercuric acetate) and endogenous stimuli (serum and RAGE ligands). Ectopic expression of the splice variant of RAGE (RAGE splice variant 4), which is resistant to ectodomain shedding, inhibited RAGE ligand dependent cell signaling, actin cytoskeleton reorganization, cell spreading, and cell migration. We found that blockade of RAGE ligand signaling with soluble RAGE or inhibitors of MAPK or PI3K blocked RAGE-dependent cell migration but did not affect RAGE splice variant 4 cell migration. We finally demonstrated that RAGE function is dependent on secretase activity as ADAM10 and γ-secretase inhibitors blocked RAGE ligand-mediated cell migration. Together, our data suggest that proteolysis of RAGE is critical to mediate signaling and cell function and may therefore emerge as a novel therapeutic target for RAGE-dependent disease states.
Collapse
Affiliation(s)
- Alex Braley
- From the Department of Cell Biology and Department of Biochemistry, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida 33136
| | - Taekyoung Kwak
- From the Department of Cell Biology and Department of Biochemistry, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida 33136
| | - Joel Jules
- From the Department of Cell Biology and Department of Biochemistry, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida 33136
| | - Evis Harja
- From the Department of Cell Biology and Department of Biochemistry, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida 33136
| | - Ralf Landgraf
- From the Department of Cell Biology and Department of Biochemistry, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida 33136
| | - Barry I Hudson
- From the Department of Cell Biology and Department of Biochemistry, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida 33136
| |
Collapse
|
18
|
Wang L, Hückelhoven A, Hong J, Jin N, Mani J, Chen BA, Schmitt M, Schmitt A. Standardization of cryopreserved peripheral blood mononuclear cells through a resting process for clinical immunomonitoring-Development of an algorithm. Cytometry A 2016; 89:246-58. [DOI: 10.1002/cyto.a.22813] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 10/18/2015] [Accepted: 12/11/2015] [Indexed: 01/05/2023]
Affiliation(s)
- Lei Wang
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Angela Hückelhoven
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Jian Hong
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Nan Jin
- Department of Hematology; Zhongda Hospital, Southeast University; Nanjing China
| | - Jiju Mani
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Bao-an Chen
- Department of Hematology; Zhongda Hospital, Southeast University; Nanjing China
| | - Michael Schmitt
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| | - Anita Schmitt
- Department of Internal Medicine V; University Clinic Heidelberg, University of Heidelberg; Germany
| |
Collapse
|
19
|
Timmerman I, Daniel AE, Kroon J, van Buul JD. Leukocytes Crossing the Endothelium: A Matter of Communication. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 322:281-329. [PMID: 26940521 DOI: 10.1016/bs.ircmb.2015.10.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Leukocytes cross the endothelial vessel wall in a process called transendothelial migration (TEM). The purpose of leukocyte TEM is to clear the causing agents of inflammation in underlying tissues, for example, bacteria and viruses. During TEM, endothelial cells initiate signals that attract and guide leukocytes to sites of tissue damage. Leukocytes react by attaching to these sites and signal their readiness to move back to endothelial cells. Endothelial cells in turn respond by facilitating the passage of leukocytes while retaining overall integrity. In this review, we present recent findings in the field and we have endeavored to synthesize a coherent picture of the intricate interplay between endothelial cells and leukocytes during TEM.
Collapse
Affiliation(s)
- Ilse Timmerman
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Anna E Daniel
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Jaap D van Buul
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands.
| |
Collapse
|
20
|
Alghamdi AS, Madill S, Foster DN, Troedsson MHT. Equine sperm-neutrophil binding. Biol Reprod 2015; 92:94. [PMID: 25695722 DOI: 10.1095/biolreprod.114.122655] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 02/16/2015] [Indexed: 11/01/2022] Open
Abstract
When mares are inseminated repeatedly, protein molecules from the seminal plasma (SP) prevent sperm-neutrophil binding and reduced fertility. The molecule(s) responsible for sperm-neutrophil binding is not known and the identification of beneficial SP proteins is complicated by their large numbers and abundant variation. We examined several important aspects of sperm-neutrophil binding to ultimately facilitate the identification and isolation of the molecule(s) responsible. First, we raised anti-equine P-selectin antibodies to determine the involvement of this adhesion molecule in sperm-neutrophil binding. While these antibodies identified equine P-selectin, they did not inhibit sperm-neutrophil binding. However, acrosome-reacted equine sperm expressed a molecule similar to the ligand recognition unit of P-selectin. Second, we attempted to characterize SP protein binding to equine sperm and gauge their affinity. Biotinylated SP proteins were incubated with fresh sperm, washed over a viscous medium, electrophoresed, and probed with avidin. Several SP proteins bound to sperm with a strong affinity to withstand these treatments. This finding may prove valuable for future attempts to identify and characterize specific SP molecules. Lastly, we compared the secretions from male sex organs/glands on sperm motility, sperm-neutrophil binding, and their protein profile. We expected fewer proteins from individual organs/glands, which would facilitate isolation and identification of target molecules. While each secretion had a varying effect on motility and sperm-neutrophil binding, the protein profile was as complex as that seen in whole SP, indicating that collection of proteins from individual sources will not facilitate this work. Together, these experiments answer several important questions related to sperm-neutrophil binding, sperm-SP proteins interaction, and the complexity of the SP proteome.
Collapse
Affiliation(s)
- Abdorrahman S Alghamdi
- Departments of Agriculture and Natural Resources, University of Minnesota Crookston, Crookston, Minnesota
| | - Scott Madill
- Veterinary Population Medicine, University of Minnesota Twin Cities, St. Paul, Minnesota
| | - Douglas N Foster
- Animal Science, University of Minnesota Twin Cities, St. Paul, Minnesota
| | - Mats H T Troedsson
- Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
21
|
Molecular mechanisms of CD8(+) T cell trafficking and localization. Cell Mol Life Sci 2015; 72:2461-73. [PMID: 25577280 DOI: 10.1007/s00018-015-1835-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 01/06/2015] [Accepted: 01/08/2015] [Indexed: 12/13/2022]
Abstract
Cytotoxic CD8(+) T cells are potent mediators of host protection against disease due to their ability to directly kill cells infected with intracellular pathogens and produce inflammatory cytokines at the site of infection. To fully achieve this objective, naïve CD8(+) T cells must be able to survey the entire body for the presence of foreign or "non-self" antigen that is delivered to draining lymph nodes following infection or tissue injury. Once activated, CD8(+) T cells undergo many rounds of cell division, acquire effector functions, and are no longer restricted to the circulation and lymphoid compartments like their naïve counterparts, but rather are drawn to inflamed tissues to combat infection. As CD8(+) T cells transition from naïve to effector to memory populations, this is accompanied by dynamic changes in the expression of adhesion molecules and chemokine receptors that ultimately dictate their localization in vivo. Thus, an understanding of the molecular mechanisms regulating CD8(+) T cell trafficking and localization is critical for vaccine design, control of infectious diseases, treatment of autoimmune disorders, and cancer immunotherapy.
Collapse
|
22
|
Gifford JL, Ishida H, Vogel HJ. Structural insights into calmodulin-regulated L-selectin ectodomain shedding. J Biol Chem 2012; 287:26513-27. [PMID: 22711531 DOI: 10.1074/jbc.m112.373373] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The L-selectin glycoprotein receptor mediates the initial steps of leukocyte migration into secondary lymphoid organs and sites of inflammation. Following cell activation through the engagement of G-protein-coupled receptors or immunoreceptors, the extracellular domains of L-selectin are rapidly shed, a process negatively controlled via the binding of the ubiquitous eukaryotic calcium-binding protein calmodulin to the cytoplasmic tail of L-selectin. Here we present the solution structure of calcium-calmodulin bound to a peptide encompassing the cytoplasmic tail and part of the transmembrane domain of L-selectin. The structure and accompanying biophysical study highlight the importance of both calcium and the transmembrane segment of L-selectin in the interaction between these two proteins, suggesting that by binding this region, calmodulin regulates in an "inside-out" fashion the ectodomain shedding of the receptor. Our structure provides the first molecular insight into the emerging new role for calmodulin as a transmembrane signaling partner.
Collapse
Affiliation(s)
- Jessica L Gifford
- Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | | | | |
Collapse
|
23
|
Cytokine production is altered in monocytes from children with hemolytic uremic syndrome. J Clin Immunol 2012; 32:622-31. [PMID: 22228570 DOI: 10.1007/s10875-011-9646-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 12/26/2011] [Indexed: 12/22/2022]
Abstract
PURPOSE The interaction of Shiga toxin (Stx) and/or lipopolysaccharide (LPS) with monocytes (Mo) may be central to the pathogenesis of hemolytic uremic syndrome (HUS), providing the cytokines necessary to sensitize endothelial cells to Stx action. We have previously demonstrated phenotypical alterations in Mo from HUS patients, including increased number of CD16+ Mo. Our aim was to investigate cytokine production in Mo from HUS patients. METHODS We evaluated TNF-α and IL-10 intracellular contents and secretion in the different Mo subsets in mild (HUS 1) and moderate/severe (HUS 2 + 3) patients. As controls, we studied healthy (HC) and infected children (IC). We also studied Mo responsive capacity towards LPS, measuring the modulation of Mo surface molecules and cytokine production. RESULTS In basal conditions, the intracellular measurement of TNF-α and IL-10 revealed that the highest number of cytokine-producing Mo was found in HUS 2 + 3 and IC, whereas LPS caused a similar increase in TNF-α and IL-10-producing Mo for all groups. However, when evaluating the release of TNF-α and IL-10, we found a diminished secretion capacity in the entire HUS group and IC compared to HC in basal and LPS conditions. Similarly, a lower Mo response to LPS in HUS 2 + 3 and IC groups was observed when surface markers were studied. CONCLUSION These results indicate that Mo from severe cases of HUS, similar to IC but different to mild HUS cases, present functional changes in Mo subpopulations and abnormal responses to LPS.
Collapse
|
24
|
Scott AJ, O'Dea KP, O'Callaghan D, Williams L, Dokpesi JO, Tatton L, Handy JM, Hogg PJ, Takata M. Reactive oxygen species and p38 mitogen-activated protein kinase mediate tumor necrosis factor α-converting enzyme (TACE/ADAM-17) activation in primary human monocytes. J Biol Chem 2011; 286:35466-35476. [PMID: 21865167 DOI: 10.1074/jbc.m111.277434] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor α-converting enzyme (TACE) is responsible for the shedding of cell surface TNF. Studies suggest that reactive oxygen species (ROS) mediate up-regulation of TACE activity by direct oxidization or modification of the protein. However, these investigations have been largely based upon nonphysiological stimulation of promonocytic cell lines which may respond and process TACE differently from primary cells. Furthermore, investigators have relied upon TACE substrate shedding as a surrogate for activity quantification. We addressed these concerns, employing a direct, cell-based fluorometric assay to investigate the regulation of TACE catalytic activity on freshly isolated primary human monocytes during LPS stimulation. We hypothesized that ROS mediate up-regulation of TACE activity indirectly, by activation of intracellular signaling pathways. LPS up-regulated TACE activity rapidly (within 30 min) without changing cell surface TACE expression. Scavenging of ROS or inhibiting their production by flavoprotein oxidoreductases significantly attenuated LPS-induced TACE activity up-regulation. Exogenous ROS (H(2)O(2)) also up-regulated TACE activity with similar kinetics and magnitude as LPS. H(2)O(2)- and LPS-induced TACE activity up-regulation were effectively abolished by a variety of selective p38 MAPK inhibitors. Activation of p38 was redox-sensitive as H(2)O(2) caused p38 phosphorylation, and ROS scavenging significantly reduced LPS-induced phospho-p38 expression. Inhibition of the p38 substrate, MAPK-activated protein kinase 2, completely attenuated TACE activity up-regulation, whereas inhibition of ERK had little effect. Lastly, inhibition of cell surface oxidoreductases prevented TACE activity up-regulation distal to p38 activation. In conclusion, our data indicate that in primary human monocytes, ROS mediate LPS-induced up-regulation of TACE activity indirectly through activation of the p38 signaling pathway.
Collapse
Affiliation(s)
- Alasdair J Scott
- Section of Anaesthetics, Pain Medicine, and Intensive Care, Faculty of Medicine, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Kieran P O'Dea
- Section of Anaesthetics, Pain Medicine, and Intensive Care, Faculty of Medicine, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - David O'Callaghan
- Section of Anaesthetics, Pain Medicine, and Intensive Care, Faculty of Medicine, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Lynn Williams
- Kennedy Institute of Rheumatology, Imperial College London, London SW7 2AZ, United Kingdom
| | - Justina O Dokpesi
- Section of Anaesthetics, Pain Medicine, and Intensive Care, Faculty of Medicine, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Louise Tatton
- Section of Anaesthetics, Pain Medicine, and Intensive Care, Faculty of Medicine, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Jonathan M Handy
- Section of Anaesthetics, Pain Medicine, and Intensive Care, Faculty of Medicine, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Philip J Hogg
- Lowy Cancer Research Centre, University of New South Wales, Sydney 2052, Australia
| | - Masao Takata
- Section of Anaesthetics, Pain Medicine, and Intensive Care, Faculty of Medicine, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom.
| |
Collapse
|
25
|
Yang S, Liu F, Wang QJ, Rosenberg SA, Morgan RA. The shedding of CD62L (L-selectin) regulates the acquisition of lytic activity in human tumor reactive T lymphocytes. PLoS One 2011; 6:e22560. [PMID: 21829468 PMCID: PMC3145643 DOI: 10.1371/journal.pone.0022560] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 06/27/2011] [Indexed: 01/13/2023] Open
Abstract
CD62L/L-selectin is a marker found on naïve T cells and further distinguishes central memory (Tcm, CD62L+) from effector memory (Tem, CD62L-) T cells. The regulation of CD62L plays a pivotal role in controlling the traffic of T lymphocytes to and from peripheral lymph nodes. CD62L is shed from the cell membrane following T cell activation, however, the physiological significance of this event remains to be elucidated. In this study, we utilized in vitro generated anti-tumor antigen T cells and melanoma lines as a model to evaluate the dynamics of CD62L shedding and expression of CD107a as a marker of lytic activity. Upon encounter, with matched tumor lines, antigen reactive T cells rapidly lose CD62L expression and this was associated with the acquisition of CD107a. By CD62L ELISA, we confirmed that this transition was mediated by the shedding of CD62L when T cells encountered specific tumor antigen. The introduction of a shedding resistant mutant of CD62L into the tumor antigen-reactive T cell line JKF6 impaired CD107a acquisition following antigen recognition and this was correlated with decreased lytic activity as measured by (51)Cr release assays. The linkage of the shedding of CD62L from the surface of anti-tumor T cells and acquisition of lytic activity, suggests a new function for CD62L in T cell effector functions and anti-tumor activity.
Collapse
Affiliation(s)
- Shicheng Yang
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Fang Liu
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Qiong J. Wang
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Steven A. Rosenberg
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Richard A. Morgan
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
26
|
Engel P, Pérez-Carmona N, Albà MM, Robertson K, Ghazal P, Angulo A. Human cytomegalovirus UL7, a homologue of the SLAM-family receptor CD229, impairs cytokine production. Immunol Cell Biol 2011; 89:753-66. [PMID: 21670740 DOI: 10.1038/icb.2011.55] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Human cytomegalovirus (HCMV), the β-herpesvirus prototype, has evolved a wide spectrum of mechanisms to counteract host immunity. Among them, HCMV uses cellular captured genes encoding molecules capable of interfering with the original host function or of fulfilling new immunomodulatory tasks. Here, we report on UL7, a novel HCMV heavily glycosylated transmembrane protein, containing an Ig-like domain that exhibits remarkable amino acid similarity to CD229, a cell-surface molecule of the signalling lymphocyte-activation molecule (SLAM) family involved in leukocyte activation. The UL7 Ig-like domain, which is well-preserved in all HCMV strains, structurally resembles the SLAM-family N-terminal Ig-variable domain responsible for the homophilic and heterophilic interactions that trigger signalling. UL7 is transcribed with early-late kinetics during the lytic infectious cycle. Using a mAb generated against the viral protein, we show that it is constitutively shed, through its mucine-like stalk, from the cell-surface. Production of soluble UL7 is enhanced by PMA and reduced by a broad-spectrum metalloproteinase inhibitor. Although UL7 does not hold the ability to interact with CD229 or other SLAM-family members, it shares with them the capacity to mediate adhesion to leukocytes, specifically to monocyte-derived DCs. Furthermore, we demonstrate that UL7 expression attenuates the production of proinflammatory cytokines TNF, IL-8 and IL-6 in DCs and myeloid cell lines. Thus, the ability of UL7 to interfere with cellular proinflammatory responses may contribute to viral persistence. These results enhance our understanding of those HCMV-encoded molecules involved in sustaining the balance between HCMV and the host immune system.
Collapse
Affiliation(s)
- Pablo Engel
- Department of Cell Biology, Immunology, and Neurosciences, Medical School, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
27
|
Deng W, Srinivasan S, Zheng X, Putkey JA, Li R. Interaction of calmodulin with L-selectin at the membrane interface: implication on the regulation of L-selectin shedding. J Mol Biol 2011; 411:220-33. [PMID: 21664913 DOI: 10.1016/j.jmb.2011.05.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 05/21/2011] [Accepted: 05/26/2011] [Indexed: 12/18/2022]
Abstract
The calmodulin (CaM) hypothesis of ectodomain shedding stipulates that CaM, an intracellular Ca²⁺-dependent regulatory protein, associates with the cytoplasmic domain of L-selectin to regulate ectodomain shedding of L-selectin on the other side of the plasma membrane. To understand the underlying molecular mechanism, we have characterized the interactions of CaM with two peptides derived from human L-selectin. The peptide ARR18 corresponds to the entire cytoplasmic domain of L-selectin (residues Ala317-Tyr334 in the mature protein), and CLS corresponds to residues Lys280-Tyr334, which contains the entire transmembrane and cytoplasmic domains of l-selectin. Monitoring the interaction by fluorescence spectroscopy and other biophysical techniques, we found that CaM can bind to ARR18 in aqueous solutions or the L-selectin cytoplasmic domain of CLS reconstituted in the phosphatidylcholine bilayer, both with an affinity of approximately 2 μM. The association is calcium independent and dynamic and involves both lobes of CaM. In a phospholipid bilayer, the positively charged L-selectin cytoplasmic domain of CLS is associated with anionic phosphatidylserine (PS) lipids at the membrane interface through electrostatic interactions. Under conditions where the PS content mimics that in the inner leaflet of the cell plasma membrane, the interaction between CaM and CLS becomes undetectable. These results suggest that the association of CaM with L-selectin in the cell can be influenced by the membrane bilayer and that anionic lipids may modulate ectodomain shedding of transmembrane receptors.
Collapse
Affiliation(s)
- Wei Deng
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
28
|
The cytoplasmic domains of TNFalpha-converting enzyme (TACE/ADAM17) and L-selectin are regulated differently by p38 MAPK and PKC to promote ectodomain shedding. Biochem J 2010; 428:293-304. [PMID: 20331435 DOI: 10.1042/bj20091611] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
L-selectin mediates the initial tethering and subsequent rolling of leucocytes along luminal walls of inflamed venules. TACE [TNFalpha (tumour necrosis factor alpha)-converting enzyme] is responsible for cleaving the membrane-proximal extracellular domain of L-selectin (also known as shedding), which reduces the efficiency of leucocyte recruitment to sites of inflammation. Many reports have highlighted roles for PKC (protein kinase C) and p38 MAPK (mitogen-activated protein kinase) in promoting L-selectin shedding with little insight into the mechanism involved. By using PMA and the phosphatase inhibitors cantharidin and calyculin A, we could selectively activate PKC or p38 MAPK respectively to promote TACE-dependent shedding of L-selectin. Interestingly, the intracellular mechanisms leading to the shedding event differed dramatically. For example, regulatory elements within the L-selectin cytoplasmic tail, such as ERM (ezrin/radixin/moesin)-binding and serine residues, were important for PKC- but not p38 MAPK-dependent shedding. Also, increased and sustained cell surface levels of TACE, and phosphorylation of its cytoplasmic tail (a hallmark of TACE activation), occurred in lymphocytes and monocytes following p38 MAPK activation. Finally, we showed that TNFalpha-induced shedding of L-selectin in monocytes was strikingly similar to cantharidin-induced shedding and suggest that this newly characterized mechanism could be physiologically relevant in inflammatory cells.
Collapse
|
29
|
Wang Y, Zhang AC, Ni Z, Herrera A, Walcheck B. ADAM17 activity and other mechanisms of soluble L-selectin production during death receptor-induced leukocyte apoptosis. THE JOURNAL OF IMMUNOLOGY 2010; 184:4447-54. [PMID: 20220092 DOI: 10.4049/jimmunol.0902925] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
L-selectin is an adhesion molecule expressed by neutrophils that broadly directs their infiltration in to sites of inflammation. It is also present at relatively high levels in the serum of normal individuals. It is well established that L-selectin is efficiently shed from the surface of neutrophils upon their activation, a process that regulates its density and binding activity. Neutrophil programmed cell death is critical for the resolution of inflammation, and L-selectin downregulation is induced during this process as well. The mechanisms underpinning this latter process are much less understood, and were investigated in this study. Using a disintegrin and metalloprotease (ADAM)-17 radiation chimeric mice, we demonstrate for the first time that during early events of death receptor-mediated neutrophil apoptosis, L-selectin downregulation occurs primarily by ADAM17-mediated shedding. This was observed as well upon using shRNA to knock down ADAM17 expression in Jurkat cells, a well-studied cell line in terms of the molecular processes involved in the induction of apoptosis. These findings directly reveal that ADAM17 activity occurs during programmed cell death. Hence, the cleavage of particular ADAM17 substrates may be an additional component of the anti-inflammatory program initiated by apoptotic neutrophils. Of interest was that during later stages of induced leukocyte apoptosis, soluble L-selectin production occurred independent of ADAM17, as well as membrane events, such as blebbing and microparticle production. This process may provide an explanation for the lack of diminished serum L-selectin levels in ADAM17-null mice, and suggests a mechanism for the homeostatic maintenance of soluble L-selectin levels in the blood of healthy individuals.
Collapse
Affiliation(s)
- Yue Wang
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St Paul, MN 55108, USA
| | | | | | | | | |
Collapse
|
30
|
Active-site determinants of substrate recognition by the metalloproteinases TACE and ADAM10. Biochem J 2009; 424:79-88. [PMID: 19715556 DOI: 10.1042/bj20090549] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The metalloproteinases TACE [tumour necrosis factor alpha-converting enzyme; also known as ADAM17 (a disintegrin and metalloproteinase 17)] and ADAM10 are the primary enzymes responsible for catalysing release of membrane-anchored proteins from the cell surface in metazoan organisms. Although the repertoire of protein substrates for these two proteases is partially overlapping, each one appears to target a subset of unique proteins in vivo. The mechanisms by which the two proteases achieve specificity for particular substrates are not completely understood. We have used peptide libraries to define the cleavage site selectivity of TACE and ADAM10. The two proteases have distinct primary sequence requirements at multiple positions surrounding the cleavage site in their substrates, which allowed us to generate peptide substrates that are highly specific for each of these proteases. The major difference between the two protease specificities maps to the P1' position (immediately downstream of the cleavage site) of the substrate. At this position, TACE is selective for smaller aliphatic residues, whereas ADAM10 can accommodate aromatic amino acids. Using mutagenesis we identified three residues in the S1' pockets of these enzymes that dramatically influence specificity for both peptide and protein substrates. Our results suggest that substrate selectivity of TACE and ADAM10 can be at least partly rationalized by specific features of their active sites.
Collapse
|
31
|
Rohrbeck A, Borlak J. Cancer genomics identifies regulatory gene networks associated with the transition from dysplasia to advanced lung adenocarcinomas induced by c-Raf-1. PLoS One 2009; 4:e7315. [PMID: 19812696 PMCID: PMC2754338 DOI: 10.1371/journal.pone.0007315] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 09/13/2009] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Lung cancer is a leading cause of cancer morbidity. To improve an understanding of molecular causes of disease a transgenic mouse model was investigated where targeted expression of the serine threonine kinase c-Raf to respiratory epithelium induced initially dysplasia and subsequently adenocarcinomas. This enables dissection of genetic events associated with precancerous and cancerous lesions. METHODOLOGY/PRINCIPAL FINDINGS By laser microdissection cancer cell populations were harvested and subjected to whole genome expression analyses. Overall 473 and 541 genes were significantly regulated, when cancer versus transgenic and non-transgenic cells were compared, giving rise to three distinct and one common regulatory gene network. At advanced stages of tumor growth predominately repression of gene expression was observed, but genes previously shown to be up-regulated in dysplasia were also up-regulated in solid tumors. Regulation of developmental programs as well as epithelial mesenchymal and mesenchymal endothelial transition was a hall mark of adenocarcinomas. Additionally, genes coding for cell adhesion, i.e. the integrins and the tight and gap junction proteins were repressed, whereas ligands for receptor tyrosine kinase such as epi- and amphiregulin were up-regulated. Notably, Vegfr- 2 and its ligand Vegfd, as well as Notch and Wnt signalling cascades were regulated as were glycosylases that influence cellular recognition. Other regulated signalling molecules included guanine exchange factors that play a role in an activation of the MAP kinases while several tumor suppressors i.e. Mcc, Hey1, Fat3, Armcx1 and Reck were significantly repressed. Finally, probable molecular switches forcing dysplastic cells into malignantly transformed cells could be identified. CONCLUSIONS/SIGNIFICANCE This study provides insight into molecular pertubations allowing dysplasia to progress further to adenocarcinoma induced by exaggerted c-Raf kinase activity.
Collapse
Affiliation(s)
- Astrid Rohrbeck
- Department of Molecular Medicine and Medical Biotechnology, Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | - Jürgen Borlak
- Department of Molecular Medicine and Medical Biotechnology, Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
- Center for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
32
|
Jia HP, Look DC, Tan P, Shi L, Hickey M, Gakhar L, Chappell MC, Wohlford-Lenane C, McCray PB. Ectodomain shedding of angiotensin converting enzyme 2 in human airway epithelia. Am J Physiol Lung Cell Mol Physiol 2009; 297:L84-96. [PMID: 19411314 DOI: 10.1152/ajplung.00071.2009] [Citation(s) in RCA: 264] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is a terminal carboxypeptidase and the receptor for the SARS and NL63 coronaviruses (CoV). Loss of ACE2 function is implicated in severe acute respiratory syndrome (SARS) pathogenesis, but little is known about ACE2 biogenesis and activity in the airways. We report that ACE2 is shed from human airway epithelia, a site of SARS-CoV infection. The regulation of ACE2 release was investigated in polarized human airway epithelia. Constitutive generation of soluble ACE2 was inhibited by DPC 333, implicating a disintegrin and metalloprotease 17 (ADAM17). Phorbol ester, ionomycin, endotoxin, and IL-1beta and TNFalpha acutely induced ACE2 release, further supporting that ADAM17 and ADAM10 regulate ACE2 cleavage. Soluble ACE2 was enzymatically active and partially inhibited virus entry into target cells. We determined that the ACE2 cleavage site resides between amino acid 716 and the putative transmembrane domain starting at amino acid 741. To reveal structural determinants underlying ACE2 release, several mutant and chimeric ACE2 proteins were engineered. Neither the juxtamembrane stalk region, transmembrane domain, nor the cytosolic domain was needed for constitutive ACE2 release. Interestingly, a point mutation in the ACE2 ectodomain, L584A, markedly attenuated shedding. The resultant ACE2-L584A mutant trafficked to the cell membrane and facilitated SARS-CoV entry into target cells, suggesting that the ACE2 ectodomain regulates its release and that residue L584 might be part of a putative sheddase "recognition motif." Thus ACE2 must be cell associated to serve as a CoV receptor and soluble ACE2 might play a role in modifying inflammatory processes at the airway mucosal surface.
Collapse
Affiliation(s)
- Hong Peng Jia
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Altered adhesion molecules expression on peripheral blood mononuclear cells from patients with systemic sclerosis and clinical correlations. Clin Rheumatol 2009; 28:847-51. [PMID: 19225705 DOI: 10.1007/s10067-009-1124-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Revised: 01/29/2009] [Accepted: 02/02/2009] [Indexed: 10/21/2022]
Abstract
The aim of the study was to evaluate the expressions of adhesion molecules (AM) on peripheral blood mononuclear cells (PBMNC) from systemic sclerosis (SSc) patients. Thirty-one SSc patients (ACR) and 20 normal subjects were selected for the study. PBMNC were analyzed for LFA-1alpha, LFA-1beta, ICAM-3, ICAM-1, and L: -selectin expressions. ICAM-3 expression was decreased while ICAM-1 was increased on SSc PBMNC, compared to controls (p = 0.04 and 0.003, respectively). A positive association was found between LFA-1alpha (r = 0.37, p = 0.03), LFA-1beta (r = 0.38, p = 0.002), ICAM-3 (r = 0.42, p = 0.01), and L-selectin (r = 0.38, p = 0.03) expressions and greater number of immunosuppressive drugs taken by SSc patients. Also, anti-centromeric positive SSc patients had lower expressions of LFA-1alpha, LFA-1beta, ICAM-3, and L-selectin. Lower expression of ICAM-3 and higher expression of ICAM-1 suggest that AMs may be involved in the pathogenesis of scleroderma.
Collapse
|
34
|
Kodera M, Grailer JJ, Karalewitz APA, Subramanian H, Steeber DA. T lymphocyte migration to lymph nodes is maintained during homeostatic proliferation. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2008; 14:211-224. [PMID: 18312727 DOI: 10.1017/s1431927608080215] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The immune system maintains appropriate cell numbers through regulation of cell proliferation and death. Normal tissue distribution of lymphocytes is maintained through expression of specific adhesion molecules and chemokine receptors such as L-selectin and CCR7, respectively. Lymphocyte insufficiency or lymphopenia induces homeostatic proliferation of existing lymphocytes to increase cell numbers. Interestingly, homeostatic proliferation of T lymphocytes induces a phenotypic change from naïve- to memory-type cell. Naïve T cells recirculate between blood and lymphoid tissues whereas memory T cells migrate to nonlymphoid sites such as skin and gut. To assess effects of homeostatic proliferation on migratory ability of T cells, a murine model of lymphopenia-induced homeostatic proliferation was used. Carboxyfluorescein diacetate, succinimidyl ester-labeled wild-type splenocytes were adoptively transferred into recombination activation gene-1-deficient mice and analyzed by flow cytometry, in vitro chemotactic and in vivo migration assays, and immunofluorescence microscopy. Homeostatically proliferated T cells acquired a mixed memory-type CD44high L-selectinhigh CCR7low phenotype. Consistent with this, chemotaxis to secondary lymphoid tissue chemokine in vitro was reduced by 22%-34%. By contrast, no differences were found for migration or entry into lymph nodes during in vivo migration assays. Therefore, T lymphocytes that have undergone homeostatic proliferation recirculate using mechanisms similar to naïve T cells.
Collapse
Affiliation(s)
- Masanari Kodera
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, USA
| | | | | | | | | |
Collapse
|
35
|
Resto VA, Burdick MM, Dagia NM, McCammon SD, Fennewald SM, Sackstein R. L-selectin-mediated lymphocyte-cancer cell interactions under low fluid shear conditions. J Biol Chem 2008; 283:15816-24. [PMID: 18385135 DOI: 10.1074/jbc.m708899200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cell migration in blood flow is mediated by engagement of specialized adhesion molecules that function under hemodynamic shear conditions, and many of the effectors of these adhesive interactions, such as the selectins and their ligands, are well defined. However, in contrast, our knowledge of the adhesion molecules operant under lymphatic flow conditions is incomplete. Among human malignancies, head and neck squamous cell cancer displays a marked predilection for locoregional lymph node metastasis. Based on this distinct tropism, we hypothesized that these cells express adhesion molecules that promote their binding to lymphoid tissue under lymphatic fluid shear stress. Accordingly, we investigated adhesive interactions between these and other cancer cells and the principal resident cells of lymphoid organs, lymphocytes. Parallel plate flow chamber studies under defined shear conditions, together with biochemical analyses, showed that human head and neck squamous cell cancer cells express heretofore unrecognized L-selectin ligand(s) that mediate binding to lymphocyte L-selectin at conspicuously low shear stress levels of 0.07-0.08 dynes/cm(2), consistent with lymphatic flow. The binding of head and neck squamous cancer cells to L-selectin displays canonical biochemical features, such as requirements for sialylation, sulfation, and N-glycosylation, but displays a novel operational shear threshold differing from all other L-selectin ligands, including those expressed on colon cancer and leukemic cells (e.g. HCELL). These data define a novel class of L-selectin ligands and expand the scope of function for L-selectin within circulatory systems to now include a novel activity within shear stresses characteristic of lymphatic flow.
Collapse
Affiliation(s)
- Vicente A Resto
- Department of Otolaryngology and the Sealy Center for Cancer Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | | | | | | | | | | |
Collapse
|
36
|
Versleijen MW, Oyen WJ, Roelofs HM, van Emst-de Vries SE, Willems PH, Jansen JB, Wanten GJ. Immune function and leukocyte sequestration under the influence of parenteral lipid emulsions in healthy humans: a placebo-controlled crossover study. Am J Clin Nutr 2008; 87:539-47. [PMID: 18326590 DOI: 10.1093/ajcn/87.3.539] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND It remains unclear whether immune modulation by lipids contributes to the high risk of infectious complications that is associated with the use of parenteral nutrition. Although mixed long- and medium-chain triacylglycerol (LCT-MCT)-containing emulsions, but not pure LCT emulsions, activate neutrophils and impair crucial leukocyte functions in vitro, in vivo studies have failed to corroborate these findings. OBJECTIVES The present investigation was conducted to evaluate the effects of LCT and LCT-MCT on immune function in healthy humans and to assess whether the lack of in vivo effects results from sampling errors due to extravascular sequestration of activated neutrophils. DESIGN Saline, LCT-MCT, and LCT emulsions were administered intravenously for 4.5 h to 12 healthy volunteers in a randomized crossover design. Plasma triacylglycerol concentrations were clamped at a clinically relevant concentration of 3-5 mmol/L. Leukocyte population counts and neutrophil activation were assessed before and after infusion. Leukocyte sequestration was evaluated by monitoring the distribution of Technetium-99m-labeled autologous leukocytes during infusions. RESULTS Whereas LCT exerted no greater effects than did saline, LCT-MCT significantly decreased lymphocyte counts. However, no evidence for neutrophil activation was found with either lipid. Moreover, the clearance of radiolabeled leukocytes from the liver, spleen, and lungs was not altered by any lipid, which suggested that lipid emulsions do not induce leukocyte sequestration. CONCLUSIONS Short-term infusion of LCT-MCT (but not LCT) to healthy humans modulates leukocyte population counts but, in clear contrast with the in vitro situation, does not induce neutrophil activation. These disparate findings cannot be explained by MCT-induced leukocyte sequestration.
Collapse
Affiliation(s)
- Michelle W Versleijen
- Department of Gastroenterology and Hepatology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands.
| | | | | | | | | | | | | |
Collapse
|
37
|
Association of Plasma Levels of F11 Receptor/Junctional Adhesion Molecule-A (F11R/JAM-A) With Human Atherosclerosis. J Am Coll Cardiol 2007; 50:1768-76. [DOI: 10.1016/j.jacc.2007.05.051] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Revised: 05/03/2007] [Accepted: 05/28/2007] [Indexed: 11/23/2022]
|
38
|
Galkina E, Florey O, Zarbock A, Smith BRE, Preece G, Lawrence MB, Haskard DO, Ager A. T lymphocyte rolling and recruitment into peripheral lymph nodes is regulated by a saturable density of L-selectin (CD62L). Eur J Immunol 2007; 37:1243-53. [PMID: 17429841 DOI: 10.1002/eji.200636481] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
L-selectin mediates tethering and rolling of lymphocytes in high endothelial venules (HEV) of lymph nodes (LN) and of leukocytes at inflammatory sites. We used transgenic mice expressing varying levels of wild-type or a non-cleavable mutant form of L-selectin on T cells to determine the relationship between L-selectin density, tethering and rolling, and migration into LN. T cells expressing supraphysiological levels of either wild-type or non-cleavable L-selectin showed rolling parameters similar to C57BL/6 T cells in hydrodynamic flow assays and during rolling in Peyer's patch HEV. In contrast, PMA- or antigen-activated T cells and L-selectin(+/-) T cells expressing subphysiological levels of L-selectin showed reduced numbers of rolling cells with increased rolling velocity. Short-term homing studies showed that elevated expression of L-selectin above physiological levels had no effect on T cell migration to LN; however, low L-selectin expression resulted in reduced T cell homing to LN. Thus, T lymphocyte migration into LN is regulated by the density of cell surface L-selectin. In addition, there is a saturable density of L-selectin required for optimal homing to PLN in C57BL/6 mice, the L-selectin level on circulating naive T cells promotes optimal homing, and increased expression above saturating levels promotes no further increase in T cell recruitment.
Collapse
Affiliation(s)
- Elena Galkina
- Division of Immunoregulation, MRC National Institute for Medical Research, The Ridgeway, London, UK.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Venturi GM, Conway RM, Steeber DA, Tedder TF. CD25+CD4+ regulatory T cell migration requires L-selectin expression: L-selectin transcriptional regulation balances constitutive receptor turnover. THE JOURNAL OF IMMUNOLOGY 2007; 178:291-300. [PMID: 17182566 DOI: 10.4049/jimmunol.178.1.291] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The molecular mechanisms controlling regulatory CD25(+)Foxp3(+)CD4(+) T cell (T(reg)) migration are central to in vivo immune responses. T(reg) cell subsets differentially express L-selectin, an adhesion molecule mediating lymphocyte migration to peripheral LNs (PLNs) and leukocyte rolling during inflammation. In this study, L-selectin was essential for T(reg) cell migration and normal tissue distribution. Specifically, there was a 90% reduction in PLN T(reg) cells in L-selectin(-/-) mice with a compensatory increase in spleen T(reg) cell numbers. Unexpectedly, however, 40% of the CD4(+) T cells remaining within PLNs of L-selectin(-/-) mice were T(reg) cells. The migratory properties of T(reg) cells were nonetheless markedly different from those of naive CD4(+) T cells, with 3- to 9-fold lower migration of T(reg) cells into PLNs and approximately 2-fold lower migration into the spleen. T(reg) cells also turned over cell surface L-selectin at a faster rate than CD25(-)CD4(+) T cells, but maintained physiologically appropriate L-selectin densities for optimal migration. Specifically, T(reg) cells expressed 30-40% more cell surface L-selectin when its endoproteolytic cleavage was blocked genetically, which resulted in a 2-fold increase in T(reg) cell migration into PLNs. However, increased L-selectin cleavage by T(reg) cells in wild-type mice was accompanied by 2-fold higher L-selectin mRNA levels, which resulted in equivalent cell surface L-selectin densities on T(reg) and naive T cells. Thus, T(reg) cells and CD25(-)CD4(+) T cells share similar requirements for L-selectin expression during migration, although additional molecular mechanisms constrain T(reg) cell migration beyond what is required for naive CD4(+) T cell migration.
Collapse
Affiliation(s)
- Guglielmo M Venturi
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
40
|
Ramos MV, Fernández GC, Patey N, Schierloh P, Exeni R, Grimoldi I, Vallejo G, Elías-Costa C, Del Carmen Sasiain M, Trachtman H, Combadière C, Proulx F, Palermo MS. Involvement of the fractalkine pathway in the pathogenesis of childhood hemolytic uremic syndrome. Blood 2006; 109:2438-45. [PMID: 17132725 DOI: 10.1182/blood-2006-06-026997] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Thrombotic microangiopathy and acute renal failure are cardinal features of postdiarrheal hemolytic uremic syndrome (HUS). These conditions are related to endothelial and epithelial cell damage induced by Shiga toxin (Stx) through the interaction with its globotriaosyl ceramide receptor. However, inflammatory processes contribute to the pathogenesis of HUS by sensitizing cells to Stx fractalkine (FKN), a CX(3)C transmembrane chemokine expressed on epithelial and endothelial cells upon activation, is involved in the selective migration and adhesion of specific leukocyte subsets to tissues. Here, we demonstrated a selective depletion of circulating mononuclear leukocytes expressing the receptor for FKN (CX(3)CR1) in patients with HUS. We found a unique phenotype in children with HUS distinct from that seen in healthy, uremic, or infected controls, in which monocytes lost CX(3)CR1, down-modulated CD62L, and increased CD16. In addition, the CD56(dim) natural killer (NK) subpopulation was decreased, leading to an altered peripheral CD56(dim)/CD56(bright) ratio from 10.0 to 4.5. It is noteworthy that a negative correlation existed between the percentage of circulating CX(3)CR1(+) leukocytes and the severity of renal failure. Finally, CX(3)CR1(+) leukocytes were observed in renal biopsies from patients with HUS. We suggest that the interaction of CX(3)CR1(+) cells with FKN present on activated endothelial cells may contribute to renal injury in HUS.
Collapse
Affiliation(s)
- María Victoria Ramos
- Division of Immunology, Institute of Hematological Investigations, Academia Nacional de Medicina, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Versleijen M, Roelofs H, Preijers F, Roos D, Wanten G. Parenteral lipids modulate leukocyte phenotypes in whole blood, depending on their fatty acid composition. Clin Nutr 2005; 24:822-9. [PMID: 15978704 DOI: 10.1016/j.clnu.2005.05.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2005] [Accepted: 05/06/2005] [Indexed: 11/17/2022]
Abstract
To characterize the immunological effects of various lipids that are applied as part of total parenteral nutrition (TPN) formulations, we analyzed phenotypical changes in leukocytes following lipid exposure. Importantly, the study was performed with whole blood in order to prevent the functional changes that are induced by isolation procedures. Briefly, blood samples from 10 healthy volunteers were incubated with lipids containing pure long-chain triglycerides (L), mixed long- and medium-chain triglycerides (LM), synthetic structured lipids (SL), or emulsions based on olive oil (OO), or fish oil (FO). After immune fluorescent staining, leukocyte phenotype characteristics were analyzed by flowcytometry. Exposure to LM increased in a dose-dependent manner the expression of membrane surface markers for adhesion (CD11b) and degranulation (CD66b), while decreasing CD62L, on neutrophils and monocytes. These findings demonstrate that LM activates leukocytes in peripheral whole blood. On the other hand, decreased expression of activation markers was observed with L and FO. Lipids effects on the phenotype of T lymphocytes and Natural Killer cells were not seen during incubation for up to 4 h. These results indicate that (i) the composition of TPN formulations with regard to lipid structure has implications for the function of exposed immune competent cells and (ii) medium-chain triglycerides, which have been regarded as functionally inert deliverers of fuel calories, have distinct biological effects.
Collapse
Affiliation(s)
- Michelle Versleijen
- Department of Gastroenterology, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
42
|
Barkhausen T, Krettek C, van Griensven M. L-selectin: adhesion, signalling and its importance in pathologic posttraumatic endotoxemia and non-septic inflammation. ACTA ACUST UNITED AC 2005; 57:39-52. [PMID: 16089318 DOI: 10.1016/j.etp.2005.02.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The leucocyte expressed surface-bound L-selectin belongs to the selectin family of adhesion molecules. It exhibits adhesive as well as signalling functions. Mainly, it is of importance in lymphocyte homing and in the extravasation of leucocytes into the surrounding tissue during inflammation. Acting in the initial step of the cell adhesion cascade, L-selectin is responsible for the rolling of leucocytes on endothelial layers. Therefore, L-selectin is thought to be an adequate target for pharmacological interventions. Beneath the discussion of the molecules' general features like molecule structure and its regulation, the review focuses firstly on L-selectin in the context of posttraumatic inflammatory disorders, and secondly on the importance of L-selectin specific signalling events.
Collapse
Affiliation(s)
- Tanja Barkhausen
- Experimental Trauma Surgery, Department of Trauma Surgery, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover, Germany.
| | | | | |
Collapse
|
43
|
Abstract
L-selectin is a cell adhesion molecule consisting of a large, highly glycosylated, extracellular domain, a single spanning transmembrane domain and a small cytoplasmic tail. It is expressed on most leukocytes and is involved in their rolling on inflamed vascular endothelium prior to firm adhesion and transmigration. It is also required for the constitutive trafficking of lymphocytes through secondary lymphoid organs. Like most adhesion molecules, L-selectin function is regulated by a variety of mechanisms including gene transcription, post-translational modifications, association with the actin cytoskeleton, and topographic distribution. In addition, it is rapidly downregulated by proteolytic cleavage near the cell surface by ADAM-17 (TACE) and at least one other "sheddase". This process of "ectodomain shedding" results in the release of most of the extracellular portion of L-selectin from the cell surface while retaining the cytoplasmic, transmembrane, and eleven amino acids of the extracellular domain on the cell. This review will examine the mechanism(s) of L-selectin ectodomain shedding and discuss the physiological implications.
Collapse
Affiliation(s)
- D M Smalley
- Cardiovascular Research Center and Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, 22908-1294, USA
| | | |
Collapse
|
44
|
Neuenburg JK, Cho TA, Nilsson A, Bredt BM, Hebert SJ, Grant RM, Price RW. T-cell activation and memory phenotypes in cerebrospinal fluid during HIV infection. J Acquir Immune Defic Syndr 2005; 39:16-22. [PMID: 15851909 DOI: 10.1097/01.qai.0000155036.03004.a0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We characterized T cell phenotypes in 74 paired blood and cerebrospinal fluid (CSF) samples of HIV-infected and uninfected persons using four-color flow cytometry. CD4+ and CD8+ T cells subsets were further characterized by identifying activated/resting and memory/naive subsets in CSF and blood using the markers CD38/HLA-DR and CD45RA/CD62L, respectively. With and without HIV-infection, the proportion of CD4+ T cells and memory T cells among T cells in CSF was higher compared to blood. In HIV-infection, activated CD4+ and CD8+ T cells in CSF were more abundant than in uninfected controls. As expected, combination antiretroviral therapy (ART) reduced T cell activation in CSF and blood.
Collapse
Affiliation(s)
- Jutta K Neuenburg
- Department of Neurology, San Francisco General Hospital, General Clinical Research Center at the University of California, San Francisco, CA 94158, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Mascarell L, Truffa-Bachi P. T lymphocyte activation initiates the degradation of the CD62L encoding mRNA and increases the transcription of the corresponding gene. Immunol Lett 2005; 94:115-22. [PMID: 15234543 DOI: 10.1016/j.imlet.2004.04.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2004] [Revised: 04/19/2004] [Accepted: 04/19/2004] [Indexed: 11/19/2022]
Abstract
Following T-cell activation, CD62L, a member of the selectin family of cell adhesion molecules, is proteolytically cleaved by a constitutive endoprotease and subsequently re-expressed. To define whether the cleavage regulates CD62L gene transcription, we have analyzed the outcome of T-cell activation on the level of CD62L gene transcription and mRNA stability. Here, we report that CD62L shedding correlates with the concomitant upregulation of CD62L gene transcription and the rapid degradation of the corresponding mRNA. Novel protein synthesis is not required for CD62L gene upregulation, mRNA degradation or protein shedding. The three events are insensitive to cyclosporin A (CSA) and, thus, do not depend on the calcineurin signaling pathway. Activation of T cells in presence of a metallo-protease inhibitor, that protects CD62L shedding, does not prevent CD62L gene upregulation or mRNA degradation. In contrast induction of CD62L shedding by the chemically-induced dissociation of calmodulin from the CD62L cytosolic tail, in absence of T-cell activation, has no consequences on the levels of CD62L gene transcription or mRNA accumulation. These data demonstrate that the transcriptional and post-transcriptional events are exclusively regulated by T-cell activation and not by the CD62L density on cell membrane.
Collapse
Affiliation(s)
- Laurent Mascarell
- Unité de Biologie des Populations Lymphocytaires, Department of Immunology, Institut Pasteur, 25 rue du Dr Roux, Cedex 15, 75724 Paris, France.
| | | |
Collapse
|
46
|
Chilton L, Middlemas A, Gardiner N, Tomlinson DR. The p75 neurotrophin receptor appears in plasma in diabetic rats-characterisation of a potential early test for neuropathy. Diabetologia 2004; 47:1924-30. [PMID: 15558233 DOI: 10.1007/s00125-004-1550-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2004] [Accepted: 07/18/2004] [Indexed: 10/26/2022]
Abstract
AIMS/HYPOTHESIS This study tested the premise that immunoreactivity representing the p75 neurotrophin receptor (p75(NTR)) appears in plasma of diabetic rats in association with the early stages of neuronal dysfunction or damage. We also examined whether treatment beneficial to neuropathy might reduce the p75(NTR) immunoreactivity. METHODS Plasma proteins were fractionated by SDS-PAGE and immunoblots exposed to p75(NTR) antibody, using receptor protein from cultured PC12 cells as an external standard. Rats were made diabetic with streptozotocin for various periods and exsanguinated. Plasma glucose, HbA(1)c and plasma proteins were determined. We also studied plasma samples from diabetic mice lacking the gene coding for p75(NTR), as well as the effect of sciatic nerve crush on healthy male Wistar rats. RESULTS Plasma p75(NTR) immunoreactivity began to exceed normal levels at 8 weeks after induction of diabetes, and was significantly raised at 10 (p<0.05) and 12 weeks (p<0.001). Treatment between 8 and 12 weeks with insulin, fidarestat (an aldose reductase inhibitor), nerve growth factor and neurotrophin 3 all normalised the plasma p75(NTR) immunoreactivity. Plasma from p75(NTR) (-/-) mice contained no such immunoreactivity, though it was present in plasma from wild-type mice. Following nerve crush, p75(NTR) immunoreactivity appeared in plasma of non-diabetic mice, indicating that this can be a result of nerve trauma. CONCLUSIONS/INTERPRETATION These observations suggest that plasma p75(NTR) immunoreactivity may serve as an early indicator of neuronal dysfunction or damage in diabetes. The time course of its appearance relates well to that of early neuropathy and its response to interventions that are neuroprotective suggests that it might mirror neurological status.
Collapse
Affiliation(s)
- L Chilton
- Division of Neuroscience, Faculty of Life Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | | | | | | |
Collapse
|
47
|
Diez-Fraile A, Meyer E, Duchateau L, Burvenich C. Effect of proinflammatory mediators and glucocorticoids on L-selectin expression in peripheral blood neutrophils from dairy cows in various stages of lactation. Am J Vet Res 2004; 65:1421-6. [PMID: 15524330 DOI: 10.2460/ajvr.2004.65.1421] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To determine whether proinflammatory mediators and glucocorticoids affect CD62L(L-selectin) expression on peripheral blood neutrophils from cows in various stages of lactation. ANIMALS 100 healthy dairy cows during early (13.1 +/- 0.79 days after parturition; n = 31), peak (58.7 +/- 1.64 days after parturition; 31), and mid (137.2 +/- 2.59 days after parturition; 38) lactation. PROCEDURE In vitro effects of relevant proinflammatory mediators that are released in response to mastitis caused by gram-negative bacteria such as lipopolysaccharide (endotoxin), tumor necrosis factor-alpha, and platelet-activating factor (PAF) on CD62L expression on bovine neutrophils were assessed by flow cytometry. Influences of cortisol and dexamethasone on CD62L expression on bovine neutrophils were also investigated. RESULTS Basal CD62L expression on neutrophils from cows during early, peak, and mid lactation were similar. Lipopolysaccharide and tumor necrosis factor-alpha had no effect on CD62L expression on neutrophils from cows at any stage of lactation. Conversely, PAF elicited a time- and dose-dependent, down regulatory effect on CD62L expression. However, no differential shedding of CD62L from neutrophils of cows at any stage of lactation were detected. In addition, no effects on CD62L expression on bovine neutrophils after whole blood incubation with cortisol or dexamethasone were observed. Incubation with glucocorticoids did not prevent the down regulatory effect of PAF on CD62L expression. CONCLUSIONS AND CLINICAL RELEVANCE Comparable basal CD62L expression on bovine neutrophils and equal amounts of CD62L shedding from bovine neutrophils during all stages of lactation suggest that variations in CD62L density are not a likely cause of susceptibility of cows to coliform-induced mastitis during early lactation.
Collapse
Affiliation(s)
- Araceli Diez-Fraile
- Department of Physiology, Biochemistry and Biometrics, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | | | | | | |
Collapse
|
48
|
Lemberg MK, Martoglio B. On the mechanism of SPP-catalysed intramembrane proteolysis; conformational control of peptide bond hydrolysis in the plane of the membrane. FEBS Lett 2004; 564:213-8. [PMID: 15111098 DOI: 10.1016/s0014-5793(04)00192-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2003] [Accepted: 02/04/2004] [Indexed: 01/01/2023]
Abstract
Intramembrane-cleaving proteases are members of a novel type of enzyme that hydrolyse substrate proteins within transmembrane regions. The presently known proteases that catalyse such cleavage reactions are membrane proteins of high hydrophobicity and multiple predicted transmembrane regions. A key feature is the positioning of active site residues in hydrophobic segments implying that the catalytic centre is assembled within the plane of the membrane. Nevertheless, all these proteases appear to utilise catalytic mechanisms similar to classic proteases that expose their active site domains in aqueous compartments. In the present review, we will address the mechanism of intramembrane proteolysis on the example of the signal peptide peptidase, and discuss how enzyme-catalysed hydrolysis of peptide bonds within the plane of a cellular membrane might occur.
Collapse
Affiliation(s)
- Marius K Lemberg
- Institute of Biochemistry, Swiss Federal Institute of Technology, ETH-Hoenggerberg, 8093 Zurich, Switzerland
| | | |
Collapse
|
49
|
Sainz IM, Uknis AB, Isordia-Salas I, Dela Cadena RA, Pixley RA, Colman RW. Interactions between bradykinin (BK) and cell adhesion molecule (CAM) expression in peptidoglycan‐polysaccharide (PG‐PS)‐induced arthritis. FASEB J 2004; 18:887-9. [PMID: 15001555 DOI: 10.1096/fj.03-0835fje] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Bradykinin (BK), a vasoactive, proinflammatory nonapeptide, promotes cell adhesion molecule (CAM) expression, leukocyte sequestration, inter-endothelial gap formation, and protein extravasation in postcapillary venules. These effects are mediated by bradykinin-1 (B1R) and-2 (B2R) receptors. We delineated some of the mechanisms by which BK could influence chronic inflammation by altering CAM expression on leukocytes, endothelium, and synovium in joint sections of peptidoglycan-polysaccharide-injected Lewis rats. Blocking B1R results in significantly increased joint inflammation. Immunohistochemistry of the B1R antagonist group revealed increased leukocyte and synovial CD11b and CD54 expression and increased CD11b and CD44 endothelial expression. B2R antagonism decreased leukocyte and synovial CD44 and CD54 and endothelial CD11b expression. Although these findings implicate B2R involvement in the acute phase of inflammation by facilitating leukocyte activation (CD11b), homing (CD44), and transmigration (CD54). Treatment with a B2R antagonist did not affect the disease evolution in this model. In contrast, when both BK receptors are blocked, the aggravation of inflammation by B1R blockade is neutralized and there is no difference from the disease-untreated model. Our findings suggest that B1R and B2R signaling show physiologic antagonism. B1R signaling suggests involvement in down-regulation of leukocyte activation, transmigration, and homing. Further studies are needed to evaluate the B1 receptor agonist's role in this model.
Collapse
Affiliation(s)
- I M Sainz
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Endothelial cells express a diverse and exquisite array of adhesion molecules and cell surface receptors. Adhesion molecules expressed on endothelial cells not only maintain structural integrity of the vasculature, but also mediate more dynamic processes such as the highly regulated movement of leukocytes from free flow into different tissue compartments. Recent studies have focused on the molecular processes that mediate endothelial cell function and their ability to respond rapidly to changes in their immediate microenvironment, as well as maintaining routine cell trafficking through specialist tissue compartments. Adhesion molecules expressed on the endothelium mediate the movement of leukocytes into the underlying extravasculature to mediate a diverse array of functions including immune effector responses, cellular interactions in specialist lymphatic microenvironments and recirculation back into the vasculature. The true diversity and capacity of adhesion molecules capable of being expressed on the endothelium is now beginning to emerge, demonstrating new levels of complexity as specialist subsets of endothelium are characterised that define specific, yet diverse functions. In this chapter, the role of cell adhesion molecules in mediating endothelial cell function is discussed, from how their different physiochemical properties contribute to function, to how specific ligand interactions expressed on leukocyte cell populations contribute to functions ranging from constitutive cell trafficking to inflammation.
Collapse
|