1
|
Thompson R, Cao X. Reassessing granzyme B: unveiling perforin-independent versatility in immune responses and therapeutic potentials. Front Immunol 2024; 15:1392535. [PMID: 38846935 PMCID: PMC11153694 DOI: 10.3389/fimmu.2024.1392535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/30/2024] [Indexed: 06/09/2024] Open
Abstract
The pivotal role of Granzyme B (GzmB) in immune responses, initially tied to cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells, has extended across diverse cell types and disease models. A number of studies have challenged conventional notions, revealing GzmB activity beyond apoptosis, impacting autoimmune diseases, inflammatory disorders, cancer, and neurotoxicity. Notably, the diverse functions of GzmB unfold through Perforin-dependent and Perforin-independent mechanisms, offering clinical implications and therapeutic insights. This review underscores the multifaceted roles of GzmB, spanning immunological and pathological contexts, which call for further investigations to pave the way for innovative targeted therapies.
Collapse
Affiliation(s)
- Raylynn Thompson
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore School of Medicine, Baltimore, MD, United States
| | - Xuefang Cao
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland Baltimore School of Medicine, Baltimore, MD, United States
| |
Collapse
|
2
|
Jusztus V, Medyouni G, Bagosi A, Lampé R, Panyi G, Matolay O, Maka E, Krasznai ZT, Vörös O, Hajdu P. Activity of Potassium Channels in CD8 + T Lymphocytes: Diagnostic and Prognostic Biomarker in Ovarian Cancer? Int J Mol Sci 2024; 25:1949. [PMID: 38396628 PMCID: PMC10888402 DOI: 10.3390/ijms25041949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/31/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
CD8+ T cells play a role in the suppression of tumor growth and immunotherapy. Ion channels control the Ca2+-dependent function of CD8+ lymphocytes such as cytokine/granzyme production and tumor killing. Kv1.3 and KCa3.1 K+ channels stabilize the negative membrane potential of T cells to maintain Ca2+ influx through CRAC channels. We assessed the expression of Kv1.3, KCa3.1 and CRAC in CD8+ cells from ovarian cancer (OC) patients (n = 7). We found that the expression level of Kv1.3 was higher in patients with malignant tumors than in control or benign tumor groups while the KCa3.1 activity was lower in the malignant tumor group as compared to the others. We demonstrated that the Ca2+ response in malignant tumor patients is higher compared to control groups. We propose that altered Kv1.3 and KCa3.1 expression in CD8+ cells in OC could be a reporter and may serve as a biomarker in diagnostics and that increased Ca2+ response through CRAC may contribute to the impaired CD8+ function.
Collapse
Affiliation(s)
- Vivien Jusztus
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (V.J.); (G.M.); (A.B.); (G.P.); (O.V.)
| | - Ghofrane Medyouni
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (V.J.); (G.M.); (A.B.); (G.P.); (O.V.)
| | - Adrienn Bagosi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (V.J.); (G.M.); (A.B.); (G.P.); (O.V.)
| | - Rudolf Lampé
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (R.L.); (O.M.); (E.M.); (Z.T.K.)
| | - György Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (V.J.); (G.M.); (A.B.); (G.P.); (O.V.)
| | - Orsolya Matolay
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (R.L.); (O.M.); (E.M.); (Z.T.K.)
| | - Eszter Maka
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (R.L.); (O.M.); (E.M.); (Z.T.K.)
| | - Zoárd Tibor Krasznai
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (R.L.); (O.M.); (E.M.); (Z.T.K.)
| | - Orsolya Vörös
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (V.J.); (G.M.); (A.B.); (G.P.); (O.V.)
| | - Péter Hajdu
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary; (V.J.); (G.M.); (A.B.); (G.P.); (O.V.)
- Division of Dental Biochemistry, Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, Egyetem tér 1., H-4032 Debrecen, Hungary
| |
Collapse
|
3
|
Ben-Eltriki M, Ahmadi AR, Nakao Y, Golla K, Lakschevitz F, Häkkinen L, Granville DJ, Kim H. Granzyme B promotes matrix metalloproteinase-1 (MMP-1) release from gingival fibroblasts in a PAR1- and Erk1/2-dependent manner: A novel role in periodontal inflammation. J Periodontal Res 2024; 59:94-103. [PMID: 37873693 DOI: 10.1111/jre.13190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/10/2023] [Accepted: 09/01/2023] [Indexed: 10/25/2023]
Abstract
OBJECTIVE To gain insights into how proteases signal to connective tissues cells in the periodontium. BACKGROUND The connective tissue degradation observed in periodontitis is largely due to matrix metalloproteinase (MMP) release by gingival fibroblasts. Granzyme B (GzmB) is a serine protease whose role in periodontitis is undefined. METHODS Human gingival crevicular fluid (GCF) samples were obtained from sites with periodontal disease and healthy control sites. GzmB was quantified in the GCF ([GzmB]GCF ) by ELISA. Gingival fibroblasts (GF) were cultured in the presence or absence of recombinant GzmB. Culture supernatants were analyzed by ELISA to quantify GzmB-induced release of interstitial collagenase (MMP-1). In some experiments, cells were pre-treated with the inhibitor PD98059 to block MEK/ERK signaling. The protease-activated receptor-1 (PAR-1) was blocked with ATAP-2 neutralizing antibody prior to GzmB stimulation. Systemic MMP-1 levels were measured in plasma from wild-type (WT) and granzyme-B-knockout (GzmB-/- ) mice. RESULTS The [GzmB]GCF in human samples was ~4-5 fold higher at sites of periodontal disease (gingivitis/periodontitis) compared to healthy control sites, suggesting an association between GzmB and localized matrix degradation. GzmB induced a ~4-5-fold increase in MMP-1 secretion by cultured fibroblasts. GzmB induced phosphorylation of Erk1/2, which was abrogated by PD98059. GzmB-induced upregulation of MMP-1 secretion was also reduced by PD98059. Blockade of PAR-1 function by ATAP-2 abrogated the increase in MMP-1 secretion by GF. Circulating MMP-1 was similar in WT and GzmB-/- mice, suggesting that GzmB's effects on MMP-1 release are not reflected systemically. CONCLUSION These data point to a novel GzmB-driven signaling pathway in fibroblasts in which MMP-1 secretion is upregulated in a PAR1- and Erk1/2-dependent manner.
Collapse
Affiliation(s)
- Mohamed Ben-Eltriki
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amir Reza Ahmadi
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yuya Nakao
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kalyan Golla
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Flavia Lakschevitz
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lari Häkkinen
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - David J Granville
- ICORD Centre and Department of Pathology and Laboratory Medicine, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hugh Kim
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
4
|
Kumagai J, Kiuchi M, Kokubo K, Yagyu H, Nemoto M, Tsuji K, Nagahata K, Sasaki A, Hishiya T, Onoue M, Shinmi R, Sonobe Y, Iinuma T, Yonekura S, Shinga J, Hanazawa T, Koseki H, Nakayama T, Yokote K, Hirahara K. The USP7-STAT3-granzyme-Par-1 axis regulates allergic inflammation by promoting differentiation of IL-5-producing Th2 cells. Proc Natl Acad Sci U S A 2023; 120:e2302903120. [PMID: 38015852 PMCID: PMC10710068 DOI: 10.1073/pnas.2302903120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 09/11/2023] [Indexed: 11/30/2023] Open
Abstract
Uncontrolled type 2 immunity by type 2 helper T (Th2) cells causes intractable allergic diseases; however, whether the interaction of CD4+ T cells shapes the pathophysiology of allergic diseases remains unclear. We identified a subset of Th2 cells that produced the serine proteases granzyme A and B early in differentiation. Granzymes cleave protease-activated receptor (Par)-1 and induce phosphorylation of p38 mitogen-activated protein kinase (MAPK), resulting in the enhanced production of IL-5 and IL-13 in both mouse and human Th2 cells. Ubiquitin-specific protease 7 (USP7) regulates IL-4-induced phosphorylation of STAT3, resulting in granzyme production during Th2 cell differentiation. Genetic deletion of Usp7 or Gzma and pharmacological blockade of granzyme B ameliorated allergic airway inflammation. Furthermore, PAR-1+ and granzyme+ Th2 cells were colocalized in nasal polyps from patients with eosinophilic chronic rhinosinusitis. Thus, the USP7-STAT3-granzymes-Par-1 pathway is a potential therapeutic target for intractable allergic diseases.
Collapse
Grants
- JP19H05650 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 20H03685 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 17K08876 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 18K07164 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 19K16683 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 21H05121 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 19K23858 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 22K15485 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP21H05120 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP20ek0410060 Japan Agency for Medical Research and Development (AMED)
- JP22ek0410092 Japan Agency for Medical Research and Development (AMED)
- JP20gm1210003 Japan Agency for Medical Research and Development (AMED)
- JPMJFR200R JST FORREST program
Collapse
Affiliation(s)
- Jin Kumagai
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Masahiro Kiuchi
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Kota Kokubo
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Hiroyuki Yagyu
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Masahiro Nemoto
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Kaori Tsuji
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Ken Nagahata
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
- Department of Rheumatology, Sapporo Medical University, Sapporo060-8556, Japan
| | - Atsushi Sasaki
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Takahisa Hishiya
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Miki Onoue
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Rie Shinmi
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Yuri Sonobe
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Tomohisa Iinuma
- Department of Otorhinolaryngology/Head and Neck Surgery, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Syuji Yonekura
- Department of Otorhinolaryngology/Head and Neck Surgery, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Jun Shinga
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa230-0045, Japan
| | - Toyoyuki Hanazawa
- Department of Otorhinolaryngology/Head and Neck Surgery, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Haruhiko Koseki
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
| | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba260-8670, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba260-8670, Japan
| |
Collapse
|
5
|
Richardson KC, Jung K, Pardo J, Turner CT, Granville DJ. Noncytotoxic Roles of Granzymes in Health and Disease. Physiology (Bethesda) 2022; 37:323-348. [PMID: 35820180 DOI: 10.1152/physiol.00011.2022] [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: 12/25/2022] Open
Abstract
Granzymes are serine proteases previously believed to play exclusive and somewhat redundant roles in lymphocyte-mediated target cell death. However, recent studies have challenged this paradigm. Distinct substrate profiles and functions have since emerged for each granzyme while their dysregulated proteolytic activities have been linked to diverse pathologies.
Collapse
Affiliation(s)
- Katlyn C Richardson
- International Collaboration on Repair Discoveries (ICORD), British Columbia Professional Firefighters' Wound Healing Laboratory, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Karen Jung
- International Collaboration on Repair Discoveries (ICORD), British Columbia Professional Firefighters' Wound Healing Laboratory, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julian Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), Zaragoza, Spain.,Department of Microbiology, Radiology, Pediatrics and Public Health, University of Zaragoza, Zaragoza, Spain.,CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Zaragoza, Spain
| | - Christopher T Turner
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia.,Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - David J Granville
- International Collaboration on Repair Discoveries (ICORD), British Columbia Professional Firefighters' Wound Healing Laboratory, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
6
|
Kaiserman D, Zhao P, Rowe CL, Leong A, Barlow N, Joeckel LT, Hitchen C, Stewart SE, Hollenberg MD, Bunnett N, Suhrbier A, Bird PI. Granzyme K initiates IL-6 and IL-8 release from epithelial cells by activating protease-activated receptor 2. PLoS One 2022; 17:e0270584. [PMID: 35881628 PMCID: PMC9321427 DOI: 10.1371/journal.pone.0270584] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/13/2022] [Indexed: 01/05/2023] Open
Abstract
Granzyme K (GzmK) is a tryptic member of the granzyme family of chymotrypsin-like serine proteases produced by cells of the immune system. Previous studies have indicated that GzmK activates protease-activated receptor 1 (PAR1) enhancing activation of monocytes and wound healing in endothelial cells. Here, we show using peptides and full length proteins that GzmK and, to a lesser extent the related protease GzmA, are capable of activating PAR1 and PAR2. These cleavage events occur at the canonical arginine P1 residue and involve exosite interactions between protease and receptor. Despite cleaving PAR2 at the same point as trypsin, GzmK does not induce a classical Ca2+ flux but instead activates a distinct signalling cascade, involving recruitment of β-arrestin and phosphorylation of ERK. In epithelial A549 cells, PAR2 activation by GzmK results in the release of inflammatory cytokines IL-6 and IL-8. These data suggest that during an immune response GzmK acts as a pro-inflammatory regulator, rather than as a cytotoxin.
Collapse
Affiliation(s)
- Dion Kaiserman
- Department of Biochemistry & Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Peishen Zhao
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia
| | - Caitlin Lorraine Rowe
- Department of Biochemistry & Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Andrea Leong
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia
| | - Nicholas Barlow
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Lars Thomas Joeckel
- Department of Biochemistry & Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Corinne Hitchen
- Department of Biochemistry & Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Sarah Elizabeth Stewart
- Department of Biochemistry & Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Morley D. Hollenberg
- Department of Physiology & Pharmacology, Department of Medicine, University of Calgary, Calgary AB, Canada
| | - Nigel Bunnett
- Department of Molecular Pathobiology, Department of Neuroscience and Physiology, Neuroscience Institute, New York University, New York, NY, United States of America
| | - Andreas Suhrbier
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Australian Infectious Disease Research Centre, University of Queensland, Brisbane, QLD, Australia
| | - Phillip Ian Bird
- Department of Biochemistry & Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
7
|
Kafai NM, Diamond MS, Fox JM. Distinct Cellular Tropism and Immune Responses to Alphavirus Infection. Annu Rev Immunol 2022; 40:615-649. [PMID: 35134315 DOI: 10.1146/annurev-immunol-101220-014952] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Alphaviruses are emerging and reemerging viruses that cause disease syndromes ranging from incapacitating arthritis to potentially fatal encephalitis. While infection by arthritogenic and encephalitic alphaviruses results in distinct clinical manifestations, both virus groups induce robust innate and adaptive immune responses. However, differences in cellular tropism, type I interferon induction, immune cell recruitment, and B and T cell responses result in differential disease progression and outcome. In this review, we discuss aspects of immune responses that contribute to protective or pathogenic outcomes after alphavirus infection. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Natasha M Kafai
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA; , .,Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Michael S Diamond
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA; , .,Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA.,Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Julie M Fox
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA;
| |
Collapse
|
8
|
Chen H, Smith M, Herz J, Li T, Hasley R, Le Saout C, Zhu Z, Cheng J, Gronda A, Martina JA, Irusta PM, Karpova T, McGavern DB, Catalfamo M. The role of protease-activated receptor 1 signaling in CD8 T cell effector functions. iScience 2021; 24:103387. [PMID: 34841225 PMCID: PMC8605340 DOI: 10.1016/j.isci.2021.103387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022] Open
Abstract
CD8 T cells are essential for adaptive immunity against viral infections. Protease activated receptor 1 (PAR1) is expressed by CD8 T cells; however, its role in T cell effector function is not well defined. Here we show that in human CD8 T cells, PAR1 stimulation accelerates calcium mobilization. Furthermore, PAR1 is involved in cytotoxic T cell function by facilitating granule trafficking via actin polymerization and repositioning of the microtubule organizing center (MTOC) toward the immunological synapse. In vivo, PAR1-/- mice have reduced cytokine-producing T cells in response to a lymphocytic choriomeningitis virus (LCMV) infection and fail to efficiently control the virus. Specific deletion of PAR1 in LCMV GP33-specific CD8 T cells results in reduced expansion and diminished effector function. These data demonstrate that PAR1 plays a role in T cell activation and function, and this pathway could represent a new therapeutic strategy to modulate CD8 T cell effector function.
Collapse
Affiliation(s)
- Hui Chen
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mindy Smith
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jasmin Herz
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Tong Li
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| | - Rebecca Hasley
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cecile Le Saout
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ziang Zhu
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| | - Jie Cheng
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| | - Andres Gronda
- Department of Human Science, Georgetown University, Washington, DC, USA
| | - José A. Martina
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pablo M. Irusta
- Department of Human Science, Georgetown University, Washington, DC, USA
| | - Tatiana Karpova
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dorian B. McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Marta Catalfamo
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| |
Collapse
|
9
|
Pompili E, De Franchis V, Giampietri C, Leone S, De Santis E, Fornai F, Fumagalli L, Fabrizi C. Protease Activated Receptor 1 and Its Ligands as Main Regulators of the Regeneration of Peripheral Nerves. Biomolecules 2021; 11:1668. [PMID: 34827666 PMCID: PMC8615415 DOI: 10.3390/biom11111668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/29/2021] [Accepted: 11/05/2021] [Indexed: 12/16/2022] Open
Abstract
In contrast with the brain and spinal cord, peripheral nerves possess a striking ability to regenerate after damage. This characteristic of the peripheral nervous system is mainly due to a specific population of glial cells, the Schwann cells. Schwann cells promptly activate after nerve injury, dedifferentiate assuming a repair phenotype, and assist axon regrowth. In general, tissue injury determines the release of a variety of proteases which, in parallel with the degradation of their specific targets, also activate plasma membrane receptors known as protease-activated receptors (PARs). PAR1, the prototypical member of the PAR family, is also known as thrombin receptor and is present at the Schwann cell plasma membrane. This receptor is emerging as a possible regulator of the pro-regenerative capacity of Schwann cells. Here, we summarize the most recent literature data describing the possible contribution of PAR1 and PAR1-activating proteases in regulating the regeneration of peripheral nerves.
Collapse
Affiliation(s)
- Elena Pompili
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy; (V.D.F.); (C.G.); (E.D.S.); (L.F.); (C.F.)
| | - Valerio De Franchis
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy; (V.D.F.); (C.G.); (E.D.S.); (L.F.); (C.F.)
| | - Claudia Giampietri
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy; (V.D.F.); (C.G.); (E.D.S.); (L.F.); (C.F.)
| | - Stefano Leone
- Department of Science, Roma Tre University, Viale Guglielmo Marconi 446, 00146 Rome, Italy;
| | - Elena De Santis
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy; (V.D.F.); (C.G.); (E.D.S.); (L.F.); (C.F.)
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| | - Lorenzo Fumagalli
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy; (V.D.F.); (C.G.); (E.D.S.); (L.F.); (C.F.)
| | - Cinzia Fabrizi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy; (V.D.F.); (C.G.); (E.D.S.); (L.F.); (C.F.)
| |
Collapse
|
10
|
Chandrabalan A, Ramachandran R. Molecular mechanisms regulating Proteinase‐Activated Receptors (PARs). FEBS J 2021; 288:2697-2726. [DOI: 10.1111/febs.15829] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Arundhasa Chandrabalan
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| |
Collapse
|
11
|
Involvement of cytotoxic Eomes-expressing CD4 + T cells in secondary progressive multiple sclerosis. Proc Natl Acad Sci U S A 2021; 118:2021818118. [PMID: 33836594 PMCID: PMC7980371 DOI: 10.1073/pnas.2021818118] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multiple sclerosis (MS), a putative autoimmune disease of the central nervous system (CNS), commonly presents as relapsing-remitting MS (RRMS), characterized by recurrent episodes of peripheral disabling symptoms resulting from inflammatory CNS damage. Many RRMS patients transition to a chronic disease course with progressive neurological dysfunctions (secondary progressive MS, SPMS), with the progression rate varying between patients and over time. SPMS pathogenesis is now linked to immune-cell-mediated processes, although the mechanisms driving SPMS transition and progression remain elusive, and SPMS lacks biomarkers and effective treatments. We report the crucial involvement of cytotoxic CD4+ T cells expressing Eomes (Eomes+ Th cells) in SPMS pathogenesis-a Th cell subset previously identified in a mouse model of late/chronic autoimmune CNS inflammation. Few Eomes+ Th cells circulate in RRMS patient peripheral blood (n = 44), primary progressive MS (PPMS) patients (n = 25), or healthy controls (n = 42), but Eomes+ Th cells were significantly increased in SPMS (n = 105, P < 0.0001). Strikingly, lymphocytes isolated from SPMS autopsy brain samples revealed CD4+ T cells infiltrating CNS that coexpressed Eomes and the cytotoxic molecule granzyme B. In particular, the Eomes+ Th cell levels were increased in SPMS patients in progressive disease phases versus SPMS patients without current disability increases (P < 0.0001). Moreover, Eomes level acted as a biomarker to predict SPMS patients at risk of disease worsening with over 80% accuracy (ROC-AUC = 0.8276). Overall, our results indicate that granzyme B-expressing Eomes+ T helper cells are involved in the pathogenesis of SPMS, with significant implications for SPMS biomarkers and therapeutic targets.
Collapse
|
12
|
Screening of small molecule libraries using combined text mining, ligand- and target-driven based approaches for identification of novel granzyme H inhibitors. J Mol Graph Model 2021; 105:107876. [PMID: 33744783 DOI: 10.1016/j.jmgm.2021.107876] [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] [Received: 11/15/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 11/23/2022]
Abstract
Granzymes are serine proteases synthesized by CTL and NK cells. Five granzyme genes (GzmA, -B, -H, -K, -M) are present in humans, which are located at three different chromosomal loci. Being serine proteases, the binding pocket constitutes a catalytic triad (i.e., His59, Asp103 and Ser197). Granzymes are released into target (cancerous and virally infected) cells by a specialized process known as granule exocytosis pathway. After internalization, these proteases initiate apoptosis. Granzymes are also involved in other non-apoptotic immune associated roles like ECM remodeling, cytokine modulation, killing of pathogens through generation of phagosomes. Their intracellular activity is regulated by specialized inhibitors knows as SERPINs. However, if these proteases are secreted in excess into the extracellular environment, their regulation becomes important as otherwise they start self-damage to the tissues thereby worsening the disease conditions. Efforts are being made to identify potential inhibitors for regulation of these proteases in an extracellular environment. Physiological and synthetic inhibitors have been reported against some members however there is no known inhibitor against extracellular human GzmH. Thus, in the current study, we investigated small molecule databases for the identification of potential molecules having the ability to inhibit GzmH by combined molecular simulations, which can ultimately be used as a potential therapeutic agent.
Collapse
|
13
|
Tang ZZ, Zhang YM, Zheng T, Huang TT, Ma TF, Liu YW. Sarsasapogenin alleviates diabetic nephropathy through suppression of chronic inflammation by down-regulating PAR-1: In vivo and in vitro study. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 78:153314. [PMID: 32882582 DOI: 10.1016/j.phymed.2020.153314] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/27/2020] [Accepted: 08/24/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Sarsasapogenin (Sar) shows good effects on diabetic nephropathy (DN) through inhibition of the NLRP3 inflammasome, yet the potential mechanism is not well known. PURPOSE This study was designed to explore the regulation of thrombin and/or its receptor protease-activated receptor 1 (PAR-1) on the NLRP3 inflammasome and NF-κB signaling in DN condition, and further expounded the molecular mechanism of Sar on DN. METHODS Streptozotocin-induced diabetic rats were treated by gavage with Sar (0, 20 and 60 mg/kg) for consecutive 10 weeks. Then urine and serum were collected for protein excretion, creatinine, urea nitrogen, and uric acid assay reflecting renal functions, renal tissue sections for periodic acid-Schiff staining and ki67 expression reflecting cell proliferation, and renal cortex for the NLRP3 inflammasome and NF-κB signaling as well as thrombin/PAR-1 signaling. High glucose-cultured human mesangial cells (HMCs) were used to further investigate the effects and mechanisms of Sar. RESULTS Sar markedly ameliorated the renal functions and mesangial cell proliferation in diabetic rats, and suppressed activation of the NLRP3 inflammasome and NF-κB in renal cortex. Moreover, Sar remarkably down-regulated PAR-1 in protein and mRNA levels but didn't affect thrombin activity in kidney, although thrombin activity was significantly decreased in the renal cortex of diabetic rats. Meanwhile, high glucose induced activation of the NLRP3 inflammasome and NF-κB, and increased PAR-1 expression while didn't change thrombin activity in HMCs; however, Sar co-treatment ameliorated all the above indices. Further studies demonstrated that PAR-1 knockdown attenuated activation of the NLRP3 inflammasome and NF-κB, and Sar addition strengthened these effects in high glucose-cultured HMCs. CONCLUSION Sar relieved DN in rat through inhibition of the NLRP3 inflammasome and NF-κB by down-regulating PAR-1 in kidney.
Collapse
Affiliation(s)
- Zhuang-Zhuang Tang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yu-Meng Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Ting Zheng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Ting-Ting Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Teng-Fei Ma
- Institute for Stem Cell and Neural Regeneration; Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yao-Wu Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Department of Pharmacology, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
14
|
Granzymes in cardiovascular injury and disease. Cell Signal 2020; 76:109804. [PMID: 33035645 DOI: 10.1016/j.cellsig.2020.109804] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 12/17/2022]
Abstract
Chronic inflammation and impaired wound healing play important roles in the pathophysiology of cardiovascular diseases. Moreover, the aberrant secretion of proteases plays a critical role in pathological tissue remodeling in chronic inflammatory conditions. Human Granzymes (Granule secreted enzymes - Gzms) comprise a family of five (GzmA, B, H, K, M) cell-secreted serine proteases. Although each unique in function and substrate specificities, Gzms were originally thought to share redundant, intracellular roles in cytotoxic lymphocyte-induced cell death. However, an abundance of evidence has challenged this dogma. It is now recognized, that individual Gzms exhibit unique substrate repertoires and functions both intracellularly and extracellularly. In the extracellular milieu, Gzms contribute to inflammation, vascular dysfunction and permeability, reduced cell adhesion, release of matrix-sequestered growth factors, receptor activation, and extracellular matrix cleavage. Despite these recent findings, the non-cytotoxic functions of Gzms in the context of cardiovascular disease pathogenesis remain poorly understood. Minimally detected in tissues and bodily fluids of normal individuals, GzmB is elevated in patients with acute coronary syndromes, coronary artery disease, and myocardial infarction. Pre-clinical animal models have exemplified the importance of GzmB in atherosclerosis, aortic aneurysm, and cardiac fibrosis as animals deficient in GzmB exhibit reduced tissue remodeling, improved disease phenotypes and increased survival. Although a role for GzmB in cardiovascular disease is described, further work to elucidate the mechanisms that underpin the remaining human Gzms activity in cardiovascular disease is necessary. The present review provides a summary of the pre-clinical and clinical evidence, as well as emerging areas of research pertaining to Gzms in tissue remodeling and cardiovascular disease.
Collapse
|
15
|
Oki S. Eomes-expressing T-helper cells as potential target of therapy in chronic neuroinflammation. Neurochem Int 2019; 130:104348. [DOI: 10.1016/j.neuint.2018.11.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 11/29/2022]
|
16
|
Ikram S, Ahmad J, Durdagi S. Screening of FDA approved drugs for finding potential inhibitors against Granzyme B as a potent drug-repurposing target. J Mol Graph Model 2019; 95:107462. [PMID: 31786094 DOI: 10.1016/j.jmgm.2019.107462] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 09/12/2019] [Accepted: 09/29/2019] [Indexed: 10/25/2022]
Abstract
Granzyme B is one of the best-characterized and extensively studied member of cytotoxic lymphocytes (CL) proteases. Initially, it is thought to be involved in eliminating virally infected or cancerous cells by using a specialized mechanism through which they are internalized into target cells. In the last decade, however this dimension has changed as there are several reports show that not only CL but also other immune cells can also synthesize Granzyme B. This leads to the possibility of the presence of these proteases in extracellular environment. Being active protease, it then raises the possibility of damaging host tissues as evident from the available reported literature. In many instances, Granzyme B is directly involved in pathogenicity, however in others, it contributes to the disease severity as their over expression makes the clinical situation quite worse which ultimately leads to the chronic state of the disease. Serine protease inhibitor-9 is a natural known intracellular inhibitor of Granzyme B, however there is less data available about the potential inhibitors that can regulate its activity in an extracellular environment. Current study is an effort to identify potential novel inhibitors of Granzyme B. For this aim, drug repurposing study was performed. Around 7900 FDA approved drugs were screened using both ligand- and target-driven approaches. Initially, all molecules were docked using induced fit docking (IFD) approach and selected 318 high-docking scored molecules were used in short (1-ns) molecular dynamics (MD) simulations. Based on MM/GBSA binding free energy calculations, 6 compounds were selected and used in long (100-ns) MD simulations. These compounds were then used in binary QSAR analysis. Therapeutic activity potentials of studied compounds were investigated by Clarivate Analytics's MetaCore/MetaDrug platform which uses binary QSAR models. It is developed based on manually curated database of molecular interactions, molecular pathways, gene-disease associations, chemical metabolism and toxicity information. Results of selected compounds were compared with a positive control molecule. Current drug repurposing study is a step ahead in finding potential lead compounds by screening database of FDA approved molecules. We have identified novel inhibitors (Tannic acid, Mupirocin, Phytonadiol sodium diphosphate, Cefpiramide, Xenazoic acid) that have potential to decrease the activity of Granzyme B.
Collapse
Affiliation(s)
- Saima Ikram
- Center of Biotechnology & Microbiology, University of Peshawar, Pakistan; Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Jamshaid Ahmad
- Center of Biotechnology & Microbiology, University of Peshawar, Pakistan.
| | - Serdar Durdagi
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey.
| |
Collapse
|
17
|
Jastrzebski S, Kalinowski J, Mun S, Shin B, Adapala NS, Jacome-Galarza CE, Mirza F, Aguila HL, Drissi H, Sanjay A, Canalis E, Lee SK, Lorenzo JA. Protease-Activated Receptor 1 Deletion Causes Enhanced Osteoclastogenesis in Response to Inflammatory Signals through a Notch2-Dependent Mechanism. THE JOURNAL OF IMMUNOLOGY 2019; 203:105-116. [PMID: 31109956 DOI: 10.4049/jimmunol.1801032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 04/25/2019] [Indexed: 12/29/2022]
Abstract
We found that protease-activated receptor 1 (PAR1) was transiently induced in cultured osteoclast precursor cells. Therefore, we examined the bone phenotype and response to resorptive stimuli of PAR1-deficient (knockout [KO]) mice. Bones and bone marrow-derived cells from PAR1 KO and wild-type (WT) mice were assessed using microcomputed tomography, histomorphometry, in vitro cultures, and RT-PCR. Osteoclastic responses to TNF-α (TNF) challenge in calvaria were analyzed with and without a specific neutralizing Ab to the Notch2-negative regulatory region (N2-NRR Ab). In vivo under homeostatic conditions, there were minimal differences in bone mass or bone cells between PAR1 KO and WT mice. However, PAR1 KO myeloid cells demonstrated enhanced osteoclastogenesis in response to receptor activator of NF-κB ligand (RANKL) or the combination of RANKL and TNF. Strikingly, in vivo osteoclastogenic responses of PAR1 KO mice to TNF were markedly enhanced. We found that N2-NRR Ab reduced TNF-induced osteoclastogenesis in PAR1 KO mice to WT levels without affecting WT responses. Similarly, in vitro N2-NRR Ab reduced RANKL-induced osteoclastogenesis in PAR1 KO cells to WT levels without altering WT responses. We conclude that PAR1 functions to limit Notch2 signaling in responses to RANKL and TNF and moderates osteoclastogenic response to these cytokines. This effect appears, at least in part, to be cell autonomous because enhanced osteoclastogenesis was seen in highly purified PAR1 KO osteoclast precursor cells. It is likely that this pathway is involved in regulating the response of bone to diseases associated with inflammatory signals.
Collapse
Affiliation(s)
| | | | - Sehwan Mun
- Center on Aging, UConn Health, Farmington, CT 06030
| | - Bongjin Shin
- Center on Aging, UConn Health, Farmington, CT 06030
| | | | | | - Faryal Mirza
- Department of Medicine, UConn Health, Farmington, CT 06030
| | | | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30329
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030
| | - Ernesto Canalis
- Department of Medicine, UConn Health, Farmington, CT 06030.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030
| | | | - Joseph A Lorenzo
- Department of Medicine, UConn Health, Farmington, CT 06030; .,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030
| |
Collapse
|
18
|
Heuberger DM, Schuepbach RA. Protease-activated receptors (PARs): mechanisms of action and potential therapeutic modulators in PAR-driven inflammatory diseases. Thromb J 2019; 17:4. [PMID: 30976204 PMCID: PMC6440139 DOI: 10.1186/s12959-019-0194-8] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/08/2019] [Indexed: 12/29/2022] Open
Abstract
Inflammatory diseases have become increasingly prevalent with industrialization. To address this, numerous anti-inflammatory agents and molecular targets have been considered in clinical trials. Among molecular targets, protease-activated receptors (PARs) are abundantly recognized for their roles in the development of chronic inflammatory diseases. In particular, several inflammatory effects are directly mediated by the sensing of proteolytic activity by PARs. PARs belong to the seven transmembrane domain G protein-coupled receptor family, but are unique in their lack of physiologically soluble ligands. In contrast with classical receptors, PARs are activated by N-terminal proteolytic cleavage. Upon removal of specific N-terminal peptides, the resulting N-termini serve as tethered activation ligands that interact with the extracellular loop 2 domain and initiate receptor signaling. In the classical pathway, activated receptors mediate signaling by recruiting G proteins. However, activation of PARs alternatively lead to the transactivation of and signaling through receptors such as co-localized PARs, ion channels, and toll-like receptors. In this review we consider PARs and their modulators as potential therapeutic agents, and summarize the current understanding of PAR functions from clinical and in vitro studies of PAR-related inflammation.
Collapse
Affiliation(s)
- Dorothea M Heuberger
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Surgical Research Division, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reto A Schuepbach
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
LoPresti P. Silent Free Fall at Disease Onset: A Perspective on Therapeutics for Progressive Multiple Sclerosis. Front Neurol 2018; 9:973. [PMID: 30542317 PMCID: PMC6277889 DOI: 10.3389/fneur.2018.00973] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/29/2018] [Indexed: 01/08/2023] Open
Abstract
Central nervous system (CNS) degeneration occurs during multiple sclerosis (MS) following several years of reversible autoimmune demyelination. Progressive CNS degeneration appears later during the course of relapsing-remitting MS (RRMS), although it starts insidiously at disease onset. We propose that there is an early subclinical phase also for primary-progressive (PP) MS. Consensus exists that many different cell types are involved during disease onset. Furthermore, the response to the initial damage, which is specific for each individual, would result in distinct pathological pathways that add complexity to the disease and the mechanisms underlying progressive CNS degeneration. Progressive MS is classified as either active or not active, as well as with or without progression. Different forms of progressive MS might reflect distinct or overlapping pathogenetic pathways. Disease mechanisms should be determined for each patient at diagnosis and the time of treatment. Until individualized and time-sensitive treatments that specifically target the molecular mechanisms of the progressive aspect of the disease are identified, combined therapies directed at anti-inflammation, regeneration, and neuroprotection are the most effective for preventing MS progression. This review presents selected therapeutics in support of the overall idea of a multidimensional therapy applied early in the disease. This approach could limit damage and increase CNS repair. By targeting several cellular populations (i.e., microglia, astrocytes, neurons, oligodendrocytes, and lymphocytes) and multiple pathological processes (e.g., inflammation, demyelination, synaptopathy, and excitatory/inhibitory imbalance) progressive MS could be attenuated. Early timing for such multidimensional therapy is proposed as the prerequisite for effectively halting progressive MS.
Collapse
Affiliation(s)
- Patrizia LoPresti
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
20
|
Human Granzyme B Based Targeted Cytolytic Fusion Proteins. Biomedicines 2018; 6:biomedicines6020072. [PMID: 29925790 PMCID: PMC6027395 DOI: 10.3390/biomedicines6020072] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/08/2018] [Accepted: 06/11/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer immunotherapy aims to selectively target and kill tumor cells whilst limiting the damage to healthy tissues. Controlled delivery of plant, bacterial and human toxins or enzymes has been shown to promote the induction of apoptosis in cancerous cells. The 4th generation of targeted effectors are being designed to be as humanized as possible—a solution to the problem of immunogenicity encountered with existing generations. Granzymes are serine proteases which naturally function in humans as integral cytolytic effectors during the programmed cell death of cancerous and pathogen-infected cells. Secreted predominantly by cytotoxic T lymphocytes and natural killer cells, granzymes function mechanistically by caspase-dependent or caspase-independent pathways. These natural characteristics make granzymes one of the most promising human enzymes for use in the development of fusion protein-based targeted therapeutic strategies for various cancers. In this review, we explore research involving the use of granzymes as cytolytic effectors fused to antibody fragments as selective binding domains.
Collapse
|
21
|
Lee PR, Johnson TP, Gnanapavan S, Giovannoni G, Wang T, Steiner JP, Medynets M, Vaal MJ, Gartner V, Nath A. Protease-activated receptor-1 activation by granzyme B causes neurotoxicity that is augmented by interleukin-1β. J Neuroinflammation 2017; 14:131. [PMID: 28655310 PMCID: PMC5488439 DOI: 10.1186/s12974-017-0901-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 06/14/2017] [Indexed: 12/05/2022] Open
Abstract
Background The cause of neurodegeneration in progressive forms of multiple sclerosis is unknown. We investigated the impact of specific neuroinflammatory markers on human neurons to identify potential therapeutic targets for neuroprotection against chronic inflammation. Methods Surface immunocytochemistry directly visualized protease-activated receptor-1 (PAR1) and interleukin-1 (IL-1) receptors on neurons in human postmortem cortex in patients with and without neuroinflammatory lesions. Viability of cultured neurons was determined after exposure to cerebrospinal fluid from patients with progressive multiple sclerosis or purified granzyme B and IL-1β. Inhibitors of PAR1 activation and of PAR1-associated second messenger signaling were used to elucidate a mechanism of neurotoxicity. Results Immunohistochemistry of human post-mortem brain tissue demonstrated cells expressing higher amounts of PAR1 near and within subcortical lesions in patients with multiple sclerosis compared to control tissue. Human cerebrospinal fluid samples containing granzyme B and IL-1β were toxic to human neuronal cultures. Granzyme B was neurotoxic through activation of PAR1 and subsequently the phospholipase Cβ-IP3 second messenger system. Inhibition of PAR1 or IP3 prevented granzyme B toxicity. IL-1β enhanced granzyme B-mediated neurotoxicity by increasing PAR1 expression. Conclusions Neurons within the inflamed central nervous system are imperiled because they express more PAR1 and are exposed to a neurotoxic combination of both granzyme B and IL-1β. The effects of these inflammatory mediators may be a contributing factor in the progressive brain atrophy associated with neuroinflammatory diseases. Knowledge of how exposure to IL-1β and granzyme B act synergistically to cause neuronal death yields potential novel neuroprotective treatments for neuroinflammatory diseases.
Collapse
Affiliation(s)
- Paul R Lee
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Building 10, Room CRC 3-2563, Bethesda, MD, 20892, USA.
| | - Tory P Johnson
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Building 10, Room CRC 3-2563, Bethesda, MD, 20892, USA
| | - Sharmilee Gnanapavan
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, UK
| | - Gavin Giovannoni
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, UK
| | - Tongguang Wang
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Building 10, Room CRC 3-2563, Bethesda, MD, 20892, USA
| | - Joseph P Steiner
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marie Medynets
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Building 10, Room CRC 3-2563, Bethesda, MD, 20892, USA
| | - Mark J Vaal
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Valerie Gartner
- Undiagnosed Diseases Program, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Building 10, Room CRC 3-2563, Bethesda, MD, 20892, USA
| |
Collapse
|
22
|
Lin C, Borensztajn K, Spek CA. Targeting coagulation factor receptors - protease-activated receptors in idiopathic pulmonary fibrosis. J Thromb Haemost 2017; 15:597-607. [PMID: 28079978 DOI: 10.1111/jth.13623] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Indexed: 12/11/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease with a 5-year mortality rate of > 50% and unknown etiology. Treatment options remain limited and, currently, only two drugs are available, i.e. nintedanib and pirfenidone. However, both of these antifibrotic agents only slow down the progression of the disease, and do not remarkably prolong the survival of IPF patients. Hence, the discovery of new therapeutic targets for IPF is crucial. Studies exploring the mechanisms that are involved in IPF have identified several possible targets for therapeutic interventions. Among these, blood coagulation factor receptors, i.e. protease-activated receptors (PARs), are key candidates, as these receptors mediate the cellular effects of coagulation factors and play central roles in influencing inflammatory and fibrotic responses. In this review, we will focus on the controversial role of the coagulation cascade in the pathogenesis of IPF. In the light of novel data, we will attempt to reconciliate the apparently conflicting data and discuss the possibility of pharmacologic targeting of PARs for the treatment of fibroproliferative diseases.
Collapse
Affiliation(s)
- C Lin
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - K Borensztajn
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
- Inserm UMR1152, Medical School Xavier Bichat, Paris, France
- Département Hospitalo-universtaire FIRE (Fibrosis, Inflammation and Remodeling) and LabEx Inflamex, Paris, France
| | - C A Spek
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
23
|
Kochenderfer JN, Somerville RPT, Lu T, Shi V, Bot A, Rossi J, Xue A, Goff SL, Yang JC, Sherry RM, Klebanoff CA, Kammula US, Sherman M, Perez A, Yuan CM, Feldman T, Friedberg JW, Roschewski MJ, Feldman SA, McIntyre L, Toomey MA, Rosenberg SA. Lymphoma Remissions Caused by Anti-CD19 Chimeric Antigen Receptor T Cells Are Associated With High Serum Interleukin-15 Levels. J Clin Oncol 2017; 35:1803-1813. [PMID: 28291388 DOI: 10.1200/jco.2016.71.3024] [Citation(s) in RCA: 442] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Purpose T cells genetically modified to express chimeric antigen receptors (CARs) targeting CD19 (CAR-19) have potent activity against acute lymphoblastic leukemia, but fewer results supporting treatment of lymphoma with CAR-19 T cells have been published. Patients with lymphoma that is chemotherapy refractory or relapsed after autologous stem-cell transplantation have a grim prognosis, and new treatments for these patients are clearly needed. Chemotherapy administered before adoptive T-cell transfer has been shown to enhance the antimalignancy activity of adoptively transferred T cells. Patients and Methods We treated 22 patients with advanced-stage lymphoma in a clinical trial of CAR-19 T cells preceded by low-dose chemotherapy. Nineteen patients had diffuse large B-cell lymphoma, two patients had follicular lymphoma, and one patient had mantle cell lymphoma. Patients received a single dose of CAR-19 T cells 2 days after a low-dose chemotherapy conditioning regimen of cyclophosphamide plus fludarabine. Results The overall remission rate was 73% with 55% complete remissions and 18% partial remissions. Eleven of 12 complete remissions are ongoing. Fifty-five percent of patients had grade 3 or 4 neurologic toxicities that completely resolved. The low-dose chemotherapy conditioning regimen depleted blood lymphocytes and increased serum interleukin-15 (IL-15). Patients who achieved a remission had a median peak blood CAR+ cell level of 98/μL and those who did not achieve a remission had a median peak blood CAR+ cell level of 15/μL ( P = .027). High serum IL-15 levels were associated with high peak blood CAR+ cell levels ( P = .001) and remissions of lymphoma ( P < .001). Conclusion CAR-19 T cells preceded by low-dose chemotherapy induced remission of advanced-stage lymphoma, and high serum IL-15 levels were associated with the effectiveness of this treatment regimen. CAR-19 T cells will likely become an important treatment for patients with relapsed lymphoma.
Collapse
Affiliation(s)
- James N Kochenderfer
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Robert P T Somerville
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Tangying Lu
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Victoria Shi
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Adrian Bot
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - John Rossi
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Allen Xue
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Stephanie L Goff
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - James C Yang
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Richard M Sherry
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Christopher A Klebanoff
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Udai S Kammula
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Marika Sherman
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Arianne Perez
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Constance M Yuan
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Tatyana Feldman
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Jonathan W Friedberg
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Mark J Roschewski
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Steven A Feldman
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Lori McIntyre
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Mary Ann Toomey
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| | - Steven A Rosenberg
- James N. Kochenderfer, Robert P.T. Somerville, Tangying Lu, Victoria Shi, Stephanie L. Goff, James C. Yang, Richard M. Sherry, Christopher A. Klebanoff, Udai S. Kammula, Constance M. Yuan, Mark J. Roschewski, Steven A. Feldman, Lori McIntyre, Mary Ann Toomey, and Steven A. Rosenberg, National Cancer Institute, National Institutes of Health, Bethesda, MD; Adrian Bot, John Rossi, Allen Xue, Marika Sherman, and Arianne Perez, Kite Pharma, Santa Monica, CA; Tatyana Feldman, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ; and Jonathan W. Friedberg, University of Rochester School of Medicine, Rochester, NY
| |
Collapse
|
24
|
Reeves TM, Trimmer PA, Colley BS, Phillips LL. Targeting Kv1.3 channels to reduce white matter pathology after traumatic brain injury. Exp Neurol 2016; 283:188-203. [PMID: 27302680 PMCID: PMC4992637 DOI: 10.1016/j.expneurol.2016.06.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/31/2016] [Accepted: 06/10/2016] [Indexed: 02/07/2023]
Abstract
Axonal injury is present in essentially all clinically significant cases of traumatic brain injury (TBI). While no effective treatment has been identified to date, experimental TBI models have shown promising axonal protection using immunosuppressants FK506 and Cyclosporine-A, with treatment benefits attributed to calcineurin inhibition or protection of mitochondrial function. However, growing evidence suggests neuroprotective efficacy of these compounds may also involve direct modulation of ion channels, and in particular Kv1.3. The present study tested whether blockade of Kv1.3 channels, using Clofazimine (CFZ), would alleviate TBI-induced white matter pathology in rodents. Postinjury CFZ administration prevented suppression of compound action potential (CAP) amplitude in the corpus callosum of adult rats following midline fluid percussion TBI, with injury and treatment effects primarily expressed in unmyelinated CAPs. Kv1.3 protein levels in callosal tissue extracts were significantly reduced postinjury, but this loss was prevented by CFZ treatment. In parallel, CFZ also attenuated the injury-induced elevation in pro-inflammatory cytokine IL1-β. The effects of CFZ on glial function were further studied using mixed microglia/astrocyte cell cultures derived from P3-5 mouse corpus callosum. Cultures of callosal glia challenged with lipopolysaccharide exhibited a dramatic increase in IL1-β levels, accompanied by reactive morphological changes in microglia, both of which were attenuated by CFZ treatment. These results support a cell specific role for Kv1.3 signaling in white matter pathology after TBI, and suggest a treatment approach based on the blockade of these channels. This therapeutic strategy may be especially efficacious for normalizing neuro-glial interactions affecting unmyelinated axons after TBI.
Collapse
Affiliation(s)
- Thomas M Reeves
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, United States
| | - Patricia A Trimmer
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, United States
| | - Beverly S Colley
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, United States
| | - Linda L Phillips
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, United States
| |
Collapse
|
25
|
Sharma M, Merkulova Y, Raithatha S, Parkinson LG, Shen Y, Cooper D, Granville DJ. Extracellular granzyme K mediates endothelial activation through the cleavage of protease-activated receptor-1. FEBS J 2016; 283:1734-47. [PMID: 26936634 DOI: 10.1111/febs.13699] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 01/25/2016] [Accepted: 02/29/2016] [Indexed: 01/26/2023]
Abstract
Granzymes are a family of serine proteases that were once thought to function exclusively as mediators of cytotoxic lymphocyte-induced target cell death. However, non-apoptotic roles for granzymes, including granzyme K (GzK), have been proposed. As recent studies have observed elevated levels of GzK in the plasma of patients diagnosed with clinical sepsis, we hypothesized that extracellular GzK induces a proinflammatory response in endothelial cells. In the present study, extracellular GzK proteolytically activated protease-activated receptor-1 leading to increased interleukin 6 and monocyte chemotactic protein 1 production in endothelial cells. Enhanced expression of intercellular adhesion molecule 1 along with an increased capacity for adherence of THP-1 cells was also observed. Characterization of downstream pathways implicated the mitogen-activated protein kinase p38 pathway for intercellular adhesion molecule 1 expression, and both the p38 and the extracellular signal-regulated protein kinases 1 and 2 pathways in cytokine production. GzK also increased tumour necrosis factor α-induced inflammatory adhesion molecule expression. Furthermore, the physiological inhibitor of GzK, inter-α-inhibitor protein, significantly inhibited GzK activity in vitro. In summary, extracellular GzK promotes a proinflammatory response in endothelial cells.
Collapse
Affiliation(s)
- Mehul Sharma
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yulia Merkulova
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Sheetal Raithatha
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada
| | - Leigh G Parkinson
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yue Shen
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Dawn Cooper
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - David J Granville
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
26
|
Lee JY, Biemond M, Petratos S. Axonal degeneration in multiple sclerosis: defining therapeutic targets by identifying the causes of pathology. Neurodegener Dis Manag 2015; 5:527-48. [DOI: 10.2217/nmt.15.50] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Current therapeutics in multiple sclerosis (MS) target the putative inflammation and immune attack on CNS myelin. Despite their effectiveness in blunting the relapse rate in MS patients, such therapeutics do not prevent MS disease progression. Importantly, specific clinical dilemma arises through inability to predict MS progression and thereby therapeutically target axonal injury during MS, limiting permanent disability. The current review identifies immune and neurobiological principles that govern the sequelae of axonal degeneration during MS disease progression. Defining the specific disease arbiters, inflammatory and autoimmune, oligodendrocyte dystrophy and degenerative myelin, we discuss a basis for a molecular mechanism in axons that may be targeted therapeutically, in spatial and temporal manner to limit axonal degeneration and thereby halt progression of MS.
Collapse
Affiliation(s)
- Jae Young Lee
- Department of Medicine, Central Clinical School, Monash University, Prahran VIC 3004, Australia
| | - Melissa Biemond
- Department of Medicine, Central Clinical School, Monash University, Prahran VIC 3004, Australia
| | - Steven Petratos
- Department of Medicine, Central Clinical School, Monash University, Prahran VIC 3004, Australia
| |
Collapse
|
27
|
Levetiracetam Prevents Perforin Mediated Neuronal Injury Induced by Acute Cerebral Ischemia Reperfusion. Mol Neurobiol 2015; 53:5480-91. [PMID: 26454821 DOI: 10.1007/s12035-015-9467-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 09/29/2015] [Indexed: 12/18/2022]
Abstract
The purpose of this study is to explore the neuroprotection mechanism of levetiracetam (LEV) with acute focal cerebral ischemia-reperfusion (I/P) mouse. The cerebral artery I/P animal model was prepared with a middle artery cerebral occlusion method. For drug intervention, mice were intraperitoneally injected with LEV with a dose of either 15 or 150 mg/kg. Neuronal injury was evaluated by measuring the infarct area, apoptosis ratio, and observation of blood-brain barrier ultrastructure with transmission electron microscope. CD8(+) antibody and perforin antibody were used to make cross-reference screen through flow cytometry to determine a perforin-positive rate in CD8(+) T lymphocytes (PFN + %). Injection of LEV can reduce infarct area, apoptosis ratio, and blood-brain barrier damage 24 h later after acute I/P in WT mice. In vitro, perforin can lower hippocampal neuron viability. In vivo, removing perforin can relieve neuronal injury. High dose injection of LEV (150 mg/kg) can inhibit perforin secreting from CD8(+)T lymphocytes. In addition, LEV can still protect neurons with perforin knockout mice. Therefore, our results suggested that LEV may contribute to neuron protection after cerebral ischemia reperfusion. The possible mechanism may be related with perforin release. However, we cannot roll out other mechanisms.
Collapse
|
28
|
Eomesodermin-expressing T-helper cells are essential for chronic neuroinflammation. Nat Commun 2015; 6:8437. [PMID: 26436530 DOI: 10.1038/ncomms9437] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/20/2015] [Indexed: 11/09/2022] Open
Abstract
Development of acute experimental autoimmune encephalomyelitis (EAE) depends on Th17 cells expressing the nuclear factor NR4A2. However, in mice lacking NR4A2 in T cells, a late-onset disease is still inducible, despite a great reduction in acute inflammation. We here reveal that development of this late onset disease depends on cytotoxic T-cell-like CD4(+) T cells expressing the T-box transcription factor Eomesodermin (Eomes). T-cell-specific deletion of the Eomes gene remarkably ameliorates the late-onset EAE. Strikingly, similar Eomes(+) CD4(+) T cells are increased in the peripheral blood and cerebrospinal fluid from patients in a progressive state of multiple sclerosis. Collective data indicate an involvement of granzyme B and protease-activated receptor-1 in the neuroinflammation mediated by Eomes(+) CD4(+) T cells.
Collapse
|
29
|
Banerji J. Asparaginase treatment side-effects may be due to genes with homopolymeric Asn codons (Review-Hypothesis). Int J Mol Med 2015; 36:607-26. [PMID: 26178806 PMCID: PMC4533780 DOI: 10.3892/ijmm.2015.2285] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/15/2015] [Indexed: 12/14/2022] Open
Abstract
The present treatment of childhood T-cell leukemias involves the systemic administration of prokary-otic L-asparaginase (ASNase), which depletes plasma Asparagine (Asn) and inhibits protein synthesis. The mechanism of therapeutic action of ASNase is poorly understood, as are the etiologies of the side-effects incurred by treatment. Protein expression from genes bearing Asn homopolymeric coding regions (N-hCR) may be particularly susceptible to Asn level fluctuation. In mammals, N-hCR are rare, short and conserved. In humans, misfunctions of genes encoding N-hCR are associated with a cluster of disorders that mimic ASNase therapy side-effects which include impaired glycemic control, dislipidemia, pancreatitis, compromised vascular integrity, and neurological dysfunction. This paper proposes that dysregulation of Asn homeostasis, potentially even by ASNase produced by the microbiome, may contribute to several clinically important syndromes by altering expression of N-hCR bearing genes. By altering amino acid abundance and modulating ribosome translocation rates at codon repeats, the microbiomic environment may contribute to genome decoding and to shaping the proteome. We suggest that impaired translation at poly Asn codons elevates diabetes risk and severity.
Collapse
Affiliation(s)
- Julian Banerji
- Center for Computational and Integrative Biology, MGH, Simches Research Center, Boston, MA 02114, USA
| |
Collapse
|
30
|
Zhou YY, Hou GQ, He SW, Xiao Z, Xu HJ, Qiu YT, Jiang S, Zheng H, Li ZY. Psora-4, a Kv1.3 Blocker, Enhances Differentiation and Maturation in Neural Progenitor Cells. CNS Neurosci Ther 2015; 21:558-67. [PMID: 25976092 DOI: 10.1111/cns.12402] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/12/2015] [Accepted: 04/14/2015] [Indexed: 01/01/2023] Open
Abstract
AIM The self-repair ability of neural progenitor cells (NPCs) has been found to be activated and protected in several therapies helpful in multiple sclerosis (MS), an inflammatory demyelinating disease of the CNS. As a potential therapeutic target in MS, the role of the ion channel Kv1.3 in NPC self-repair has received limited attention. The aim of this study was to explore the effects of a selective Kv1.3 blocker on NPC neuronal differentiation and maturation. METHODS A small-molecule selective blocker for Kv1.3, Psora-4, was added to the differentiation medium of cultured mouse NPCs to assess its effect on NPC differentiation efficiency. Both a polypeptide Kv1.3 blocker and Kv1.3-specific RNA interference were used in parallel experiments. Further, the maturity of newborn neurons in the presence of Psora-4 was measured both by morphological analysis and by whole-cell patch clamping. RESULTS Psora-4 induced a significant increase in the percentage of neurons. Knockdown of Kv1.3 in NPCs also promoted neuronal differentiation. Both morphological and electrophysiological analyses suggested that NPC-derived neurons in the presence of Psora-4 were more mature. CONCLUSION Our studies reveal a crucial role for the ion channel Kv1.3 in the regulation of NPC differentiation and maturation, making Psora-4 a promising candidate molecule for MS treatment.
Collapse
Affiliation(s)
- Yu-Ye Zhou
- Key Laboratory of Regenerative Biology, South China Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Guo-Qiang Hou
- Key Laboratory of Regenerative Biology, South China Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Song-Wei He
- Key Laboratory of Regenerative Biology, South China Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Zhuo Xiao
- Key Laboratory of Regenerative Biology, South China Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Hui-Juan Xu
- Key Laboratory of Regenerative Biology, South China Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Ya-Tao Qiu
- Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Sheng Jiang
- Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Hui Zheng
- Key Laboratory of Regenerative Biology, South China Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Zhi-Yuan Li
- Key Laboratory of Regenerative Biology, South China Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.,Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|
31
|
Chang SC, Huq R, Chhabra S, Beeton C, Pennington MW, Smith BJ, Norton RS. N-Terminally extended analogues of the K⁺ channel toxin from Stichodactyla helianthus as potent and selective blockers of the voltage-gated potassium channel Kv1.3. FEBS J 2015; 282:2247-59. [PMID: 25864722 DOI: 10.1111/febs.13294] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 03/22/2015] [Accepted: 04/06/2015] [Indexed: 12/20/2022]
Abstract
The voltage-gated potassium channel Kv1.3 is an important target for the treatment of autoimmune diseases and asthma. Blockade of Kv1.3 by the sea anemone peptide K⁺-channel toxin from Stichodactyla helianthus (ShK) inhibits the proliferation of effector memory T lymphocytes and ameliorates autoimmune diseases in animal models. However, the lack of selectivity of ShK for Kv1.3 over the Kv1.1 subtype has driven a search for Kv1.3-selective analogues. In the present study, we describe N-terminally extended analogues of ShK that contain a negatively-charged Glu, designed to mimic the phosphonate adduct in earlier Kv1.3-selective analogues, and consist entirely of common protein amino acids. Molecular dynamics simulations indicated that a Trp residue at position [-3] of the tetrapeptide extension could form stable interactions with Pro377 of Kv1.3 and best discriminates between Kv1.3 and Kv1.1. This led to the development of ShK with an N-terminal Glu-Trp-Ser-Ser extension ([EWSS]ShK), which inhibits Kv1.3 with an IC₅₀ of 34 pm and is 158-fold selective for Kv1.3 over Kv1.1. In addition, [EWSS]ShK is more than 2900-fold more selective for Kv1.3 over Kv1.2 and KCa3.1 channels. As a highly Kv1.3-selective analogue of ShK based entirely on protein amino acids, which can be produced by recombinant expression, this peptide is a valuable addition to the complement of therapeutic candidates for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Shih C Chang
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic., Australia
| | - Redwan Huq
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Sandeep Chhabra
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic., Australia
| | - Christine Beeton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | | | - Brian J Smith
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Vic., Australia
| | - Raymond S Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic., Australia
| |
Collapse
|
32
|
Steiner JP, Bachani M, Wolfson-Stofko B, Lee MH, Wang T, Li G, Li W, Strayer D, Haughey NJ, Nath A. Interaction of paroxetine with mitochondrial proteins mediates neuroprotection. Neurotherapeutics 2015; 12:200-16. [PMID: 25404050 PMCID: PMC4322069 DOI: 10.1007/s13311-014-0315-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
There are severe neurological complications that arise from HIV infection, ranging from peripheral sensory neuropathy to cognitive decline and dementia for which no specific treatments are available. The HIV proteins secreted from infected macrophages, gp120 and Tat, are neurotoxic. The goal of this study was to screen, identify and develop neuroprotective compounds relevant to HIV-associated neurocognitive disorders (HAND). We screened more than 2000 compounds that included FDA approved drugs for protective efficacy against oxidative stress-mediated neurodegeneration and identified selective serotonin reuptake inhibitors (SSRIs) as potential neuroprotectants. Numerous SSRIs were then extensively evaluated as protectants against neurotoxicity as measured by changes in neuronal cell death, mitochondrial potential, and axodendritic degeneration elicited by HIV Tat and gp120 and other mitochondrial toxins. While many SSRIs demonstrated neuroprotective actions, paroxetine was potently neuroprotective (100 nM potency) against these toxins in vitro and in vivo following systemic administration in a gp120 neurotoxicity model. Interestingly, the inhibition of serotonin reuptake by paroxetine was not required for neuroprotection, since depletion of the serotonin transporter had no effect on its neuroprotective properties. We determined that paroxetine interacts selectively and preferentially with brain mitochondrial proteins and blocks calcium-dependent swelling but had less effect on liver mitochondria. Additionally, paroxetine induced proliferation of neural progenitor cells in vitro and in vivo in gp120 transgenic animals. Therefore, SSRIs such as paroxetine may provide a novel adjunctive neuroprotective and neuroregenerative therapy to treat HIV-infected individuals.
Collapse
Affiliation(s)
- Joseph P. Steiner
- />Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
- />Translational Neuroscience Center, National Institute of Neurological Diseases and Stroke, National Bldg 10, Room 7C-105, 10 Center Drive, Bethesda, MD 20892 USA
| | - Muznabanu Bachani
- />Translational Neuroscience Center, National Institute of Neurological Diseases and Stroke, National Bldg 10, Room 7C-105, 10 Center Drive, Bethesda, MD 20892 USA
| | - Brett Wolfson-Stofko
- />Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
| | - Myoung-Hwa Lee
- />Section of Infections of the Nervous System, National Institute of Neurological Diseases and Stroke, National Institute of Health, Bldg 10, Room 7C103, 10 Center Drive, Bethesda, MD 20892 USA
| | - Tonguang Wang
- />Translational Neuroscience Center, National Institute of Neurological Diseases and Stroke, National Bldg 10, Room 7C-105, 10 Center Drive, Bethesda, MD 20892 USA
| | - Guanhan Li
- />Section of Infections of the Nervous System, National Institute of Neurological Diseases and Stroke, National Institute of Health, Bldg 10, Room 7C103, 10 Center Drive, Bethesda, MD 20892 USA
| | - Wenxue Li
- />Section of Infections of the Nervous System, National Institute of Neurological Diseases and Stroke, National Institute of Health, Bldg 10, Room 7C103, 10 Center Drive, Bethesda, MD 20892 USA
| | - David Strayer
- />Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Norman J. Haughey
- />Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
| | - Avindra Nath
- />Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
- />Translational Neuroscience Center, National Institute of Neurological Diseases and Stroke, National Bldg 10, Room 7C-105, 10 Center Drive, Bethesda, MD 20892 USA
- />Section of Infections of the Nervous System, National Institute of Neurological Diseases and Stroke, National Institute of Health, Bldg 10, Room 7C103, 10 Center Drive, Bethesda, MD 20892 USA
| |
Collapse
|
33
|
Kulcsar KA, Baxter VK, Greene IP, Griffin DE. Interleukin 10 modulation of pathogenic Th17 cells during fatal alphavirus encephalomyelitis. Proc Natl Acad Sci U S A 2014; 111:16053-8. [PMID: 25362048 PMCID: PMC4234572 DOI: 10.1073/pnas.1418966111] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mosquito-borne alphaviruses are important causes of epidemic encephalomyelitis. Neuronal cell death during fatal alphavirus encephalomyelitis is immune-mediated; however, the types of cells involved and their regulation have not been determined. We show that the virus-induced inflammatory response was accompanied by production of the regulatory cytokine IL-10, and in the absence of IL-10, paralytic disease occurred earlier and mice died faster. To determine the reason for accelerated disease in the absence of IL-10, immune responses in the CNS of IL-10(-/-) and wild-type (WT) mice were compared. There were no differences in the amounts of brain inflammation or peak virus replication; however, IL-10(-/-) animals had accelerated and increased infiltration of CD4(+)IL-17A(+) and CD4(+)IL-17A(+)IFNγ(+) cells compared with WT animals. Th17 cells infiltrating the brain demonstrated a pathogenic phenotype with the expression of the transcription factor, Tbet, and the production of granzyme B, IL-22, and GM-CSF, with greater production of GM-CSF in IL-10(-/-) mice. Therefore, in fatal alphavirus encephalomyelitis, pathogenic Th17 cells enter the CNS at the onset of neurologic disease and, in the absence of IL-10, appear earlier, develop into Th1/Th17 cells more often, and have greater production of GM-CSF. This study demonstrates a role for pathogenic Th17 cells in fatal viral encephalitis.
Collapse
Affiliation(s)
- Kirsten A Kulcsar
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | - Victoria K Baxter
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | - Ivorlyne P Greene
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| | - Diane E Griffin
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
| |
Collapse
|
34
|
Nath A. Eradication of human immunodeficiency virus from brain reservoirs. J Neurovirol 2014; 21:227-34. [PMID: 25366659 DOI: 10.1007/s13365-014-0291-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 09/10/2014] [Accepted: 09/18/2014] [Indexed: 11/24/2022]
Abstract
Isolated cases in which human immunodeficiency virus (HIV) infection was claimed to have been eradicated generated renewed interest in HIV reservoirs in the brain particularly since attempts to reproduce the findings using genetically engineered stem cells and immune- or myeloablation have failed. A clear understanding of the cell types in which the virus resides in the brain, the mechanism of viral persistence, restricted replication and latency, and the turnover rate of the infected cells is critical for us to develop ways to control or get rid of the virus in the brain. The brain has several unique features compared to other reservoirs. There are no resident T cells in the brain; the virus resides in macrophages and astrocytes where the viral infection is non-cytopathic. The virus evolves in the brain and since the turnover rate of these cells is low, the virus has the potential to reside in these cells for several decades and possibly for the life of the individual. This review discusses the HIV reservoirs in the brain, issues related to eradication of the virus from sanctuaries in the brain, and current challenges faced by neuroscientists in finding a cure.
Collapse
Affiliation(s)
- Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological disorders and Stroke, National Institutes of Health, Bldg 10/ 7C-103, 10 Center Drive, Bethesda, MD, 20892, USA,
| |
Collapse
|
35
|
Joeckel LT, Bird PI. Are all granzymes cytotoxic in vivo? Biol Chem 2014; 395:181-202. [DOI: 10.1515/hsz-2013-0238] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 08/30/2013] [Indexed: 01/01/2023]
Abstract
Abstract
Granzymes are serine proteases mainly found in cytotoxic lymphocytes. The most-studied member of this group is granzyme B, which is a potent cytotoxin that has set the paradigm that all granzymes are cyototoxic. In the last 5 years, this paradigm has become controversial. On one hand, there is a plethora of sometimes contradictory publications showing mainly caspase-independent cytotoxic effects of granzyme A and the so-called orphan granzymes in vitro. On the other hand, there are increasing numbers of reports of granzymes failing to induce cell death in vitro unless very high (potentially supra-physiological) concentrations are used. Furthermore, experiments with granzyme A or granzyme M knock-out mice reveal little or no deficit in their cytotoxic lymphocytes’ killing ability ex vivo, but indicate impairment in the inflammatory response. These findings of non-cytotoxic effects of granzymes challenge dogma, and thus require alternative or additional explanations to be developed of the role of granzymes in defeating pathogens. Here we review evidence for granzyme cytotoxicity, give an overview of their non-cytotoxic functions, and suggest technical improvements for future investigations.
Collapse
|
36
|
Abstract
Neurocognitive disorders are a feared complication of HIV infection, especially in the post-antiretroviral era as patients are living longer. These disorders are challenging in terms of diagnosis and treatment. The clinical syndrome has evolved, driven in part by comorbidities such as aging, drug abuse, psychiatric illnesses, and a metabolic syndrome associated with the use of antiretroviral drugs. Additionally some individuals may develop a fulminant immune reconstitution syndrome. Hence, treatment of these patients needs to be individualized. The focus of research in the HIV field has recently switched towards elimination of the HIV reservoir as a means of combating long-term HIV complications. However, these approaches may be suitable for limited populations and might not be applicable once the HIV reservoir has been established in the brain. Further, all clinical trials using neuroprotective or anti-inflammatory drugs for treatment of HIV-associated neurocognitive disorders have been unsuccessful. Hence, neurological complications of HIV infection are the biggest challenge facing HIV researchers, and there is a critical need to develop new diagnostics and approaches for treatment of these disorders.
Collapse
Affiliation(s)
- Tariq B Alfahad
- Section of Infections of the Nervous System, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
37
|
|
38
|
Hiebert PR, Granville DJ. Granzyme B in injury, inflammation, and repair. Trends Mol Med 2012; 18:732-41. [DOI: 10.1016/j.molmed.2012.09.009] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 09/14/2012] [Accepted: 09/27/2012] [Indexed: 01/24/2023]
|