1
|
Islam H, Jackson GS, Boultbee J, Tsai SH, Moreno-Cabañas A, Teixeira AADS, Wright DC, Mui AL, Little JP. Interleukin-10 resistance in type 2 diabetes is associated with defective STAT3 signaling in human blood leukocytes. Am J Physiol Cell Physiol 2025; 328:C1293-C1302. [PMID: 40080435 DOI: 10.1152/ajpcell.00124.2025] [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: 02/10/2025] [Revised: 02/24/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
Chronic inflammation is strongly implicated in the pathophysiology of type 2 diabetes (T2D), highlighting the need to better understand inflammatory processes in people living with T2D. Hyperglycemia blunts the anti-inflammatory actions of interleukin-10 (IL-10)-the most potent anti-inflammatory cytokine-but the mechanistic basis remains unclear. To test the hypothesis that signaling defects underpin this hyporesponsiveness to IL-10 action, fasted blood samples were obtained from individuals living with T2D (n = 17, age: 64 ± 9 yr, HbA1c: 7.2 ± 1.1%) and their age-matched counterparts without diabetes (n = 19, 65 ± 8 yr, 5.5 ± 0.3%). Blood leukocytes were analyzed for IL-10-mediated signaling, gene expression, and cytokine secretion using flow cytometry, qPCR, and whole blood cultures, respectively. Despite no overt elevations in circulating pro- and anti-inflammatory cytokine concentrations, blood leukocytes from individuals with T2D exhibited exaggerated cytokine secretion when exposed to lipopolysaccharide (LPS) (P < 0.05). IL-10's ability to activate its canonical transcription factor signal transducer and activator of transcription 3 (STAT3) was blunted in CD14 monocytes and CD4 lymphocytes from people with T2D (P < 0.01)-a defect associated with lower IL-10 receptor expression on both cell types (P < 0.05). This upstream signaling defect was accompanied by attenuated suppressor of cytokine signaling 3 mRNA levels in IL-10-treated mononuclear cells (P = 0.059) and higher lipopolysaccharide (LPS)-stimulated cytokine secretion from blood leukocytes exposed to IL-10 (P < 0.01). Our findings identify defective IL-10-mediated signaling and gene expression as a potential mechanism underpinning IL-10 resistance in T2D, highlighting the need for further investigation into therapeutic approaches targeting IL-10.NEW & NOTEWORTHY Our findings demonstrate that immune cells from people with type 2 diabetes (T2D) are less responsive to the anti-inflammatory actions of interleukin-10 (IL-10), which may drive chronic inflammation in this population. We identify T2D-associated defects at multiple steps of the IL-10 cascade-including IL-10 receptor expression, STAT3 signaling, SOCS3 mRNA, and cytokine secretion. Our findings highlight defective IL-10 action as a potential therapeutic target to ameliorate inflammation in T2D.
Collapse
Affiliation(s)
- Hashim Islam
- School of Health and Exercise Sciences, The University of British Columbia, Kelowna, British Columbia, Canada
| | - Garett S Jackson
- School of Health and Exercise Sciences, The University of British Columbia, Kelowna, British Columbia, Canada
| | - Jordan Boultbee
- School of Health and Exercise Sciences, The University of British Columbia, Kelowna, British Columbia, Canada
| | - Shun-Hsi Tsai
- Office of Physical Education, National Taipei University, New Taipei City, Taiwan
| | | | | | - David C Wright
- School of Kinesiology, The University of British Columbia, Vancouver, British Columbia, Canada
- Faculty of Land and Food Systems, The University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Alice L Mui
- Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan P Little
- School of Health and Exercise Sciences, The University of British Columbia, Kelowna, British Columbia, Canada
| |
Collapse
|
2
|
Solleti SK, Matthews BE, Wu J, Rowe RK. SHIP-1 Differentially Regulates IgE-Induced IL-10 and Antiviral Responses in Human Monocytes. Eur J Immunol 2025; 55:e202451065. [PMID: 39668409 DOI: 10.1002/eji.202451065] [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: 02/12/2024] [Revised: 11/20/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024]
Abstract
IgE-mediated stimulation of monocytes regulates multiple cellular functions including cellular maturation, cytokine release, antiviral responses, and T-cell differentiation. Expression of the high-affinity IgE receptor, FcεRI, is closely linked to serum IgE levels and atopic disease. The signaling molecules regulating FcεRI effector functions have been well studied in mast cells and basophils; however, less is known about the signaling and regulatory mechanisms in monocytes. This study sought to identify regulators of IgE-mediated cytokine release in human monocytes. SHIP-1 was identified as a negative regulator of IgE-induced IL-10 production. It was also determined that IgE-mediated stimulation and SHIP-1 inhibition decreased antiviral IP-10 production after liposomal poly(I:C) stimulation, indicating differential regulation by SHIP-1 in IgE-driven and antiviral response pathways. SHIP-1 and NF-κB were activated following IgE-mediated stimulation of monocytes, and NF-κB activation was related to both SHIP-1 and FcεRIα cellular expression levels. To our knowledge, this is the first study to identify a role for SHIP-1 in regulating IgE-mediated and antiviral responses in human monocytes. Given the importance of monocytes in inflammation and immune responses, a better understanding of the signaling and regulatory mechanisms downstream of the FcεRI receptor could lead to new therapeutic targets in allergic disease.
Collapse
Affiliation(s)
- Siva Kumar Solleti
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Bailey E Matthews
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Jingyi Wu
- Biomedical Genetics and Genomics Graduate Program, University of Rochester Medical Center, Rochester, New York, USA
| | - Regina K Rowe
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
3
|
Müller SM, Nelson N, Jücker M. Functional Characterization of the SHIP1-Domains Regarding Their Contribution to Inositol 5-Phosphatase Activity. Biomolecules 2025; 15:105. [PMID: 39858499 PMCID: PMC11763786 DOI: 10.3390/biom15010105] [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: 11/29/2024] [Revised: 12/18/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The Src homology 2 domain-containing inositol 5-phosphatase 1 (SHIP1) is a multidomain protein consisting of two protein-protein interaction domains, the Src homology 2 (SH2) domain, and the proline-rich region (PRR), as well as three phosphoinositide-binding domains, the pleckstrin homology-like (PHL) domain, the 5-phosphatase (5PPase) domain, and the C2 domain. SHIP1 is commonly known for its involvement in the regulation of the PI3K/AKT signaling pathway by dephosphorylation of phosphatidylinositol-3,4,5-trisphosphate (PtdIns(3,4,5)P3) at the D5 position of the inositol ring. However, the functional role of each domain of SHIP1 for the regulation of its enzymatic activity is not well understood. To determine the contribution of the individual domains to catalytic activity, the full-length protein was compared with truncated constructs lacking one or more domain(s), regarding the substrate turnover (kcat) and catalytic efficiency (kcat/Km) towards ci8-PtdIns(3,4,5)P3. With this approach, it was possible to verify the allosteric activation of SHIP1 mediated by the C2 domain as described previously, while the PHL domain seemed instead to have a negative effect regarding catalytic efficiency. The full-length SHIP1 clearly displayed the highest turnover and the second-highest catalytic efficiency, showing the role of the SH2 domain and PRR not only in protein-protein interactions but also in catalysis. The SH2 domain increased substrate turnover but negatively affected catalytic efficiency. The linker between the SH2 and the PHL domains decreased the turnover number but positively influenced the catalytic efficiency. The PRR increased both the substrate turnover and the protein's catalytic efficiency. The regression analysis of the Michaelis-Menten graph revealed SHIP1 to be an allosteric enzyme, with the PRR and the linker being the most involved domains in that regard. In summary, our data indicate a complex regulation of the enzymatic activity of SHIP1 by its individual domains. While the C2 domain and PRR at the carboxy-terminus have a positive effect on enzymatic activity, the SH2 and PHL domain at the amino-terminus inhibit catalytic efficiency.
Collapse
Affiliation(s)
| | | | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (S.M.M.); (N.N.)
| |
Collapse
|
4
|
Islam H, Tsai SH, Figueiredo C, Jackson GS, Marcotte-Chénard A, Bosak J, Moreno-Cabañas A, Lira FS, Little JP. Direct assessment of leukocyte signalling and cytokine secretion reveals exercise intensity-dependent reductions in anti-inflammatory cytokine action. J Physiol 2024; 602:2717-2736. [PMID: 38776176 DOI: 10.1113/jp286228] [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: 01/04/2024] [Accepted: 04/24/2024] [Indexed: 05/24/2024] Open
Abstract
Circulating interleukin (IL)-6 and IL-10 concentrations are widely used to evaluate the anti-inflammatory effects of exercise but do not capture cytokine action at the cellular level. Whether and how acute exercise impacts anti-inflammatory cytokine action in humans is unknown. To determine how exercise intensity and pattern impact IL-6 and IL-10 action in blood leukocytes, 16 active adults (eight males/eight females; age: 30 ± 3 years; body mass index: 22.8 ± 2.3 kg/m2;V ̇ O 2 peak ${{\dot{V}}_{{{{\mathrm{O}}}_{\mathrm{2}}}{\mathrm{peak}}}}$ : 51 ± 6 mL/kg/min) completed a no-exercise control condition (CTL) or isocaloric bouts of cycling performed below (moderate continuous exercise; MCE) or above (heavy continuous or heavy intermittent exercise; HCE or HIE, respectively) lactate threshold. Venous blood (before, after, 30 min after and 90 min after exercise) was analysed for immune cell subpopulations, plasma cytokine concentrations, anti-inflammatory cytokine action and monocyte phenotype. Exercise induced rapid leukocytosis (P < 0.001) and increased plasma IL-6 (P < 0.001), IL-10 (P = 0.0145) and tumour necrosis factor-⍺ (TNF-⍺) (P = 0.0338) concentrations in an intensity-dependent manner (HCE and/or HIE vs. CTL). These systemic changes coincided with a diminished ability of IL-10/6 to phosphorylate STAT3 (P < 0.001) and inhibit TNF-⍺ secretion (P = 0.0238) in blood leukocytes following HCE and HIE. Monocyte polarization experiments revealed lower CD80 [MCE (P = 0.0933) and HIE (P = 0.0187) vs. CTL] and a tendency for higher CD163 expression (HCE vs. CTL, P = 0.0985), suggesting that hyporesponsiveness to anti-inflammatory cytokine action does not impede the ability of exercise to promote an anti-inflammatory monocyte phenotype. These findings provide novel insights into the immunomodulatory effects of exercise in humans and highlight the importance of directly measuring cellular cytokine action when evaluating the anti-inflammatory effects of exercise. KEY POINTS: Circulating cytokine concentrations are frequently used to evaluate the anti-inflammatory effects of exercise but may not capture changes in cytokine action occurring at the cellular level. We directly assessed anti-inflammatory cytokine action - measured using a combination of intracellular signalling and cytokine secretion ex vivo - in distinct immune cell subpopulations after acute calorie-matched exercise bouts differing in intensity and pattern. Anti-inflammatory cytokine action was blunted following higher intensity exercise despite corresponding increases in circulating cytokine concentrations and immune cell counts. Changes in cytokine action were not explained by changes in cytokine receptor expression on circulating immune cells. Our findings provide new insights into the immunomodulatory effects of exercise in humans and highlight the importance of directly measuring cellular cytokine action when evaluating the anti-inflammatory effects of exercise.
Collapse
Affiliation(s)
- Hashim Islam
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Shun-Hsi Tsai
- Department of Physical Education and Sport Sciences, National Taiwan Normal University, Taipei, Taiwan
| | - Caíque Figueiredo
- Exercise and Immunometabolism Research Group, Department of Physical Education, Post-Graduation Program in Movement Sciences, State University of São Paulo (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Garett S Jackson
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Alexis Marcotte-Chénard
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Johannes Bosak
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | | | - Fabio S Lira
- Exercise and Immunometabolism Research Group, Department of Physical Education, Post-Graduation Program in Movement Sciences, State University of São Paulo (UNESP), Presidente Prudente, São Paulo, Brazil
- Centro de Investigação em Desporto e Atividade Física, University of Coimbra, Coimbra, Portugal
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| |
Collapse
|
5
|
Bradshaw WJ, Kennedy EC, Moreira T, Smith LA, Chalk R, Katis VL, Benesch JLP, Brennan PE, Murphy EJ, Gileadi O. Regulation of inositol 5-phosphatase activity by the C2 domain of SHIP1 and SHIP2. Structure 2024; 32:453-466.e6. [PMID: 38309262 PMCID: PMC10997489 DOI: 10.1016/j.str.2024.01.005] [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: 08/01/2023] [Revised: 12/12/2023] [Accepted: 01/08/2024] [Indexed: 02/05/2024]
Abstract
SHIP1, an inositol 5-phosphatase, plays a central role in cellular signaling. As such, it has been implicated in many conditions. Exploiting SHIP1 as a drug target will require structural knowledge and the design of selective small molecules. We have determined apo, and magnesium and phosphate-bound structures of the phosphatase and C2 domains of SHIP1. The C2 domains of SHIP1 and the related SHIP2 modulate the activity of the phosphatase domain. To understand the mechanism, we performed activity assays, hydrogen-deuterium exchange mass spectrometry, and molecular dynamics on SHIP1 and SHIP2. Our findings demonstrate that the influence of the C2 domain is more pronounced for SHIP2 than SHIP1. We determined 91 structures of SHIP1 with fragments bound, with some near the interface between the two domains. We performed a mass spectrometry screen and determined four structures with covalent fragments. These structures could act as starting points for the development of potent, selective probes.
Collapse
Affiliation(s)
- William J Bradshaw
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK.
| | - Emma C Kennedy
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK
| | - Tiago Moreira
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK
| | - Luke A Smith
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Rod Chalk
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK
| | - Vittorio L Katis
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK
| | - Justin L P Benesch
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Paul E Brennan
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK
| | - Emma J Murphy
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK
| | - Opher Gileadi
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK.
| |
Collapse
|
6
|
Al-Qahtani AA, Alhamlan FS, Al-Qahtani AA. Pro-Inflammatory and Anti-Inflammatory Interleukins in Infectious Diseases: A Comprehensive Review. Trop Med Infect Dis 2024; 9:13. [PMID: 38251210 PMCID: PMC10818686 DOI: 10.3390/tropicalmed9010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/29/2023] [Accepted: 12/10/2023] [Indexed: 01/23/2024] Open
Abstract
Interleukins (ILs) are signaling molecules that are crucial in regulating immune responses during infectious diseases. Pro-inflammatory ILs contribute to the activation and recruitment of immune cells, whereas anti-inflammatory ILs help to suppress excessive inflammation and promote tissue repair. Here, we provide a comprehensive overview of the role of pro-inflammatory and anti-inflammatory ILs in infectious diseases, with a focus on the mechanisms underlying their effects, their diagnostic and therapeutic potential, and emerging trends in IL-based therapies.
Collapse
Affiliation(s)
- Arwa A. Al-Qahtani
- Department of Family Medicine, College of Medicine, Al-Imam Mohammad Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia;
| | - Fatimah S. Alhamlan
- Department of Infection and Immunity, King Faisal Specialist Hospital & Research Center, Riyadh 11211, Saudi Arabia;
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| | - Ahmed Ali Al-Qahtani
- Department of Infection and Immunity, King Faisal Specialist Hospital & Research Center, Riyadh 11211, Saudi Arabia;
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| |
Collapse
|
7
|
Meyer ST, Fernandes S, Anderson RE, Pacherille A, Toms B, Kerr WG, Chisholm JD. Structure-Activity Studies on Bis-Sulfonamide SHIP1 Activators. Molecules 2023; 28:8048. [PMID: 38138538 PMCID: PMC10745928 DOI: 10.3390/molecules28248048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
The SH2-containing inositol polyphosphate 5-phosphatase 1 (SHIP1) enzyme opposes the activity of PI3K and therefore is of interest in the treatment of inflammatory disorders. Recent results also indicate that SHIP1 promotes phagolysosomal degradation of lipids by microglia, suggesting that the enzyme may be a target for the treatment of Alzheimer's disease. Therefore, small molecules that increase SHIP1 activity may have benefits in these areas. Recently we discovered a bis-sulfonamide that increases the enzymatic activity of SHIP1. A series of similar SHIP1 activators have been synthesized and evaluated to determine structure-activity relationships and improve in vivo stability. Some new analogs have now been found with improved potency. In addition, both the thiophene and the thiomorpholine in the parent structure can be replaced by groups without a low valent sulfur atom, which provides a way to access activators that are less prone to oxidative degradation.
Collapse
Affiliation(s)
- Shea T. Meyer
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA
| | - Sandra Fernandes
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | | | - Angela Pacherille
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA
| | - Bonnie Toms
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - William G. Kerr
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - John D. Chisholm
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA
| |
Collapse
|
8
|
Yeoh WJ, Krebs P. SHIP1 and its role for innate immune regulation-Novel targets for immunotherapy. Eur J Immunol 2023; 53:e2350446. [PMID: 37742135 DOI: 10.1002/eji.202350446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/03/2023] [Accepted: 09/21/2023] [Indexed: 09/25/2023]
Abstract
Phosphoinositide-3-kinase/AKT (PI3K/AKT) signaling plays key roles in the regulation of cellular activity in both health and disease. In immune cells, this PI3K/AKT pathway is critically regulated by the phosphoinositide phosphatase SHIP1, which has been reported to modulate the function of most immune subsets. In this review, we summarize our current knowledge of SHIP1 with a focus on innate immune cells, where we reflect on the most pertinent aspects described in the current literature. We also present several small-molecule agonists and antagonists of SHIP1 developed over the last two decades, which have led to improved outcomes in several preclinical models of disease. We outline these promising findings and put them in relation to human diseases with unmet medical needs, where we discuss the most attractive targets for immune therapies based on SHIP1 modulation.
Collapse
Affiliation(s)
- Wen Jie Yeoh
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Philippe Krebs
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
9
|
Terzioglu G, Young-Pearse TL. Microglial function, INPP5D/SHIP1 signaling, and NLRP3 inflammasome activation: implications for Alzheimer's disease. Mol Neurodegener 2023; 18:89. [PMID: 38017562 PMCID: PMC10685641 DOI: 10.1186/s13024-023-00674-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 10/26/2023] [Indexed: 11/30/2023] Open
Abstract
Recent genetic studies on Alzheimer's disease (AD) have brought microglia under the spotlight, as loci associated with AD risk are enriched in genes expressed in microglia. Several of these genes have been recognized for their central roles in microglial functions. Increasing evidence suggests that SHIP1, the protein encoded by the AD-associated gene INPP5D, is an important regulator of microglial phagocytosis and immune response. A recent study from our group identified SHIP1 as a negative regulator of the NLRP3 inflammasome in human iPSC-derived microglial cells (iMGs). In addition, we found evidence for a connection between SHIP1 activity and inflammasome activation in the AD brain. The NLRP3 inflammasome is a multiprotein complex that induces the secretion of pro-inflammatory cytokines as part of innate immune responses against pathogens and endogenous damage signals. Previously published studies have suggested that the NLRP3 inflammasome is activated in AD and contributes to AD-related pathology. Here, we provide an overview of the current understanding of the microglial NLRP3 inflammasome in the context of AD-related inflammation. We then review the known intracellular functions of SHIP1, including its role in phosphoinositide signaling, interactions with microglial phagocytic receptors such as TREM2 and evidence for its intersection with NLRP3 inflammasome signaling. Through rigorous examination of the intricate connections between microglial signaling pathways across several experimental systems and postmortem analyses, the field will be better equipped to tailor newly emerging therapeutic strategies targeting microglia in neurodegenerative diseases.
Collapse
Affiliation(s)
- Gizem Terzioglu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Rd, Boston, MA, 02115, USA
| | - Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Rd, Boston, MA, 02115, USA.
| |
Collapse
|
10
|
Mopuru R. IL-10 Dysregulation in Bipolar Disorder: Potential Mechanisms and Treatment Implications. Indian J Psychol Med 2023; 45:439. [PMID: 37483581 PMCID: PMC10357918 DOI: 10.1177/02537176231154270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/25/2023] Open
Affiliation(s)
- Ranesh Mopuru
- Centre for Fundamental Research and Creative Education, Bengaluru, Karnataka, India
| |
Collapse
|
11
|
Gelemanović A, Ćatipović Ardalić T, Pribisalić A, Hayward C, Kolčić I, Polašek O. Genome-Wide Meta-Analysis Identifies Multiple Novel Rare Variants to Predict Common Human Infectious Diseases Risk. Int J Mol Sci 2023; 24:7006. [PMID: 37108169 PMCID: PMC10138356 DOI: 10.3390/ijms24087006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/02/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Infectious diseases still threaten global human health, and host genetic factors have been indicated as determining risk factors for observed variations in disease susceptibility, severity, and outcome. We performed a genome-wide meta-analysis on 4624 subjects from the 10,001 Dalmatians cohort, with 14 infection-related traits. Despite a rather small number of cases in some instances, we detected 29 infection-related genetic associations, mostly belonging to rare variants. Notably, the list included the genes CD28, INPP5D, ITPKB, MACROD2, and RSF1, all of which have known roles in the immune response. Expanding our knowledge on rare variants could contribute to the development of genetic panels that could assist in predicting an individual's life-long susceptibility to major infectious diseases. In addition, longitudinal biobanks are an interesting source of information for identifying the host genetic variants involved in infectious disease susceptibility and severity. Since infectious diseases continue to act as a selective pressure on our genomes, there is a constant need for a large consortium of biobanks with access to genetic and environmental data to further elucidate the complex mechanisms behind host-pathogen interactions and infectious disease susceptibility.
Collapse
Affiliation(s)
- Andrea Gelemanović
- Department of Public Health, University of Split School of Medicine, 21000 Split, Croatia
| | | | - Ajka Pribisalić
- Department of Public Health, University of Split School of Medicine, 21000 Split, Croatia
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Ivana Kolčić
- Department of Public Health, University of Split School of Medicine, 21000 Split, Croatia
- Department of General Courses, Algebra University College, 10000 Zagreb, Croatia
| | - Ozren Polašek
- Department of Public Health, University of Split School of Medicine, 21000 Split, Croatia
- Department of General Courses, Algebra University College, 10000 Zagreb, Croatia
| |
Collapse
|
12
|
Bartolo ND, Mortimer N, Manter MA, Sanchez N, Riley M, O'Malley TT, Hooker JM. Identification and Prioritization of PET Neuroimaging Targets for Microglial Phenotypes Associated with Microglial Activity in Alzheimer's Disease. ACS Chem Neurosci 2022; 13:3641-3660. [PMID: 36473177 DOI: 10.1021/acschemneuro.2c00607] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Activation of microglial cells accompanies the progression of many neurodegenerative disorders, including Alzheimer's disease (AD). Development of molecular imaging tools specific to microglia can help elucidate the mechanism through which microglia contribute to the pathogenesis and progression of neurodegenerative disorders. Through analysis of published genetic, transcriptomic, and proteomic data sets, we identified 19 genes with microglia-specific expression that we then ranked based on association with the AD characteristics, change in expression, and potential druggability of the target. We believe that the process we used to identify and rank microglia-specific genes is broadly applicable to the identification and evaluation of targets in other disease areas and for applications beyond molecular imaging.
Collapse
Affiliation(s)
- Nicole D Bartolo
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| | - Niall Mortimer
- Human Biology and Data Science, Eisai Center for Genetics Guided Dementia Discovery, 35 Cambridgepark Drive, Cambridge, Massachusetts 02140, United States
| | - Mariah A Manter
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| | - Nicholas Sanchez
- Human Biology and Data Science, Eisai Center for Genetics Guided Dementia Discovery, 35 Cambridgepark Drive, Cambridge, Massachusetts 02140, United States
| | - Misha Riley
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| | - Tiernan T O'Malley
- Human Biology and Data Science, Eisai Center for Genetics Guided Dementia Discovery, 35 Cambridgepark Drive, Cambridge, Massachusetts 02140, United States
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
13
|
Ruffolo G, Alfano V, Romagnolo A, Zimmer T, Mills JD, Cifelli P, Gaeta A, Morano A, Anink J, Mühlebner A, Vezzani A, Aronica E, Palma E. GABA A receptor function is enhanced by Interleukin-10 in human epileptogenic gangliogliomas and its effect is counteracted by Interleukin-1β. Sci Rep 2022; 12:17956. [PMID: 36289354 PMCID: PMC9605959 DOI: 10.1038/s41598-022-22806-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/19/2022] [Indexed: 01/24/2023] Open
Abstract
Gangliogliomas (GGs) are low-grade brain tumours that cause intractable focal epilepsy in children and adults. In GG, as in epileptogenic focal malformations (i.e., tuberous sclerosis complex, TSC), there is evidence of sustained neuroinflammation with involvement of the pro-inflammatory cytokine IL-1β. On the other hand, anti-inflammatory mediators are less studied but bear relevance for understanding seizure mechanisms. Therefore, we investigated the effect of the key anti-inflammatory cytokine IL-10 on GABAergic neurotransmission in GG. We assessed the IL-10 dependent signaling by transcriptomic analysis, immunohistochemistry and performed voltage-clamp recordings on Xenopus oocytes microtransplanted with cell membranes from brain specimens, to overcome the limited availability of acute GG slices. We report that IL-10-related mRNAs were up-regulated in GG and slightly in TSC. Moreover, we found IL-10 receptors are expressed by neurons and astroglia. Furthermore, GABA currents were potentiated significantly by IL-10 in GG. This effect was time and dose-dependent and inhibited by blockade of IL-10 signaling. Notably, in the same tissue, IL-1β reduced GABA current amplitude and prevented the IL-10 effect. These results suggest that in epileptogenic tissue, pro-inflammatory mechanisms of hyperexcitability prevail over key anti-inflammatory pathways enhancing GABAergic inhibition. Hence, boosting the effects of specific anti-inflammatory molecules could resolve inflammation and reduce intractable seizures.
Collapse
Affiliation(s)
- Gabriele Ruffolo
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy ,grid.18887.3e0000000417581884IRCCS San Raffaele Roma, Rome, Italy
| | - Veronica Alfano
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy ,grid.18887.3e0000000417581884IRCCS San Raffaele Roma, Rome, Italy
| | - Alessia Romagnolo
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands
| | - Till Zimmer
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands
| | - James D. Mills
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands ,grid.83440.3b0000000121901201Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK ,grid.452379.e0000 0004 0386 7187Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | - Pierangelo Cifelli
- grid.158820.60000 0004 1757 2611Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, L’Aquila, Italy
| | - Alessandro Gaeta
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy
| | - Alessandra Morano
- grid.7841.aDepartment of Human Neuroscience, University of Rome Sapienza, Rome, Italy
| | - Jasper Anink
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands
| | - Angelika Mühlebner
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands ,grid.7692.a0000000090126352Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Annamaria Vezzani
- grid.4527.40000000106678902Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Eleonora Aronica
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands ,grid.419298.f0000 0004 0631 9143Stichting Epilepsie Instellingen Nederland, Heemstede, The Netherlands
| | - Eleonora Palma
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy ,grid.18887.3e0000000417581884IRCCS San Raffaele Roma, Rome, Italy
| |
Collapse
|
14
|
Dungan OM, Dormann S, Fernandes S, Duffy BC, Effiong DG, Kerr WG, Chisholm JD. Synthetic studies on the indane SHIP1 agonist AQX-1125. Org Biomol Chem 2022; 20:4016-4020. [PMID: 35506893 DOI: 10.1039/d2ob00555g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
AQX-1125 is an indane based SHIP1 agonist that has been evaluated in the clinic for the treatment of bladder pain syndrome/interstitial cystitis. To support our own studies on SHIP1 agonists as potential treatments for IBD and Crohn's disease, a new synthetic route to the SHIP1 agonist AQX-1125 has been developed. This sequence utilizes a hydroxy-acid intermediate which allows for ready differentiation of the C6 and C7 positions. The role of the C17 alkene in the biological activity of the system is also investigated, and this functional group is not required for SHIP1 agonist activity. While AQX-1125 shows SHIP1 agonist activity in enzyme assays, it does not show activity in cell based assays similar to other SHIP1 agonists, which limits the utility of this molecule.
Collapse
Affiliation(s)
- Otto M Dungan
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| | - Shawn Dormann
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| | - Sandra Fernandes
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Brian C Duffy
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| | - Daniel G Effiong
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| | - William G Kerr
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA. .,Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.,Department of Pediatrics, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - John D Chisholm
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| |
Collapse
|
15
|
Islam H, Jackson GS, Yoon JSJ, Cabral-Santos C, Lira FS, Mui AL, Little JP. Sex differences in IL-10's anti-inflammatory function: Greater STAT3 activation and stronger inhibition of TNF-α production in male blood leukocytes ex vivo. Am J Physiol Cell Physiol 2022; 322:C1095-C1104. [PMID: 35508192 DOI: 10.1152/ajpcell.00091.2022] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Interleukin-10 (IL-10) inhibits pro-inflammatory cytokine production in blood leukocytes - an effect mediated by signal transducer and activator of transcription 3 (STAT3) activation. To examine potential sex-based differences in IL-10's anti-inflammatory function, we treated whole blood from healthy males and females (n=16 each; age: 28±6 years; body mass index: 23.5±2.3 kg/m2) with increasing concentrations of IL-10 (1-100 ng/mL) and quantified changes in phosphorylated STAT3 (pSTAT3) in CD14+ monocytes and CD4+ lymphocytes via flow cytometry. In parallel, liposaccharide (LPS)-stimulated whole-blood cultures were used to assess sex-based differences in IL-10's ability to inhibit tumour necrosis factor (TNF)-α production. IL-10 concentration-dependently increased pSTAT3 mean fluorescent intensity (MFI) in CD14+ and CD4+ cells (main effects of concentration, P<0.01) with males exhibiting larger changes in pSTAT3 MFI in both cell types (main effects of sex, P<0.01). Accordingly, IL-10-mediated inhibition of TNF-α production was more pronounced in males (main effect of sex, P<0.01) with changes in other monocyte-derived cytokines (IL-1b, IL-1RA, IL-15) also supporting a sexual dimorphism in IL-10 action (P<0.05). These sex-based differences were not explained by differences in circulating plasma IL-10 concentrations, basal IL-10 receptor expression in unstimulated CD14+ and CD4+ cells, nor the basal expression of IL-10 signaling proteins (STAT3, SHIP1, p38 MAPK) in unstimulated peripheral blood mononuclear cells. We conclude that IL-10's anti-inflammatory function differs between male and female blood leukocytes ex vivo. This sexual dimorphism should be considered in future work investigating IL-10's anti-inflammatory action in humans as it may represent a mechanism contributing to sex differences in overall immune function.
Collapse
Affiliation(s)
- Hashim Islam
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Garett S Jackson
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Jeff S J Yoon
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Carolina Cabral-Santos
- Exercise and Immunometabolism Research Group, Department of Physical Education, Post-Graduation Program in Movement Sciences, State University of São Paulo (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Fabio S Lira
- Exercise and Immunometabolism Research Group, Department of Physical Education, Post-Graduation Program in Movement Sciences, State University of São Paulo (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Alice L Mui
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| |
Collapse
|
16
|
Pedicone C, Fernandes S, Matera A, Meyer ST, Loh S, Ha JH, Bernard D, Chisholm JD, Paolicelli RC, Kerr WG. Discovery of a novel SHIP1 agonist that promotes degradation of lipid-laden phagocytic cargo by microglia. iScience 2022; 25:104170. [PMID: 35465359 PMCID: PMC9020084 DOI: 10.1016/j.isci.2022.104170] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/31/2021] [Accepted: 03/24/2022] [Indexed: 12/01/2022] Open
Abstract
Here, we describe the use of artificial intelligence to identify novel agonists of the SH2-containing 5′ inositol phosphatase 1 (SHIP1). One of the compounds, K306, represents the most potent agonist identified to date. We find that K306 exhibits selectivity for SHIP1 vs. the paralog enzyme SHIP2, and this activation does not require the C2 domain of SHIP1 which other known SHIP1 agonists require. Thus, K306 represents a new class of SHIP1 agonists with a novel mode of agonism. Importantly, we find that K306 can suppress induction of inflammatory cytokines and iNOS in macrophages or microglia, but not by their SHIP1-deficient counterparts. K306 also reduces TNF-α production in vivo in an LPS-induced endotoxemia assay. Finally, we show that K306 enhances phagolysosomal degradation of synaptosomes and dead neurons by microglia revealing a novel function for SHIP1 that might be exploited therapeutically in dementia. Discovery of a potent SHIP1 selective agonist (K306) via artificial intelligence SHIP1 agonism via K306 is independent of the C2 domain and increases PI(3,4)P2 levels K306 reduces IL-6, TNF-α, and iNOS induction in microglia and macrophages K306 promotes phagocytic degradation of lipid-laden but not protein cargo in microglia
Collapse
Affiliation(s)
- Chiara Pedicone
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Sandra Fernandes
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Alessandro Matera
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Shea T Meyer
- Chemistry Department, Syracuse University, Syracuse, NY 13210, USA
| | - Stewart Loh
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Jeung-Hoi Ha
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | | | - John D Chisholm
- Chemistry Department, Syracuse University, Syracuse, NY 13210, USA
| | | | - William G Kerr
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.,Chemistry Department, Syracuse University, Syracuse, NY 13210, USA
| |
Collapse
|
17
|
Immunosuppressive Signaling Pathways as Targeted Cancer Therapies. Biomedicines 2022. [DOI: 10.3390/biomedicines10030682
expr 829797163 + 949875436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Immune response has been shown to play an important role in defining patient prognosis and response to cancer treatment. Tumor-induced immunosuppression encouraged the recent development of new chemotherapeutic agents that assists in the augmentation of immune responses. Molecular mechanisms that tumors use to evade immunosurveillance are attributed to their ability to alter antigen processing/presentation pathways and the tumor microenvironment. Cancer cells take advantage of normal molecular and immunoregulatory machinery to survive and thrive. Cancer cells constantly adjust their genetic makeup using several mechanisms such as nucleotide excision repair as well as microsatellite and chromosomal instability, thus giving rise to new variants with reduced immunogenicity and the ability to continue to grow without restrictions. This review will focus on the central molecular signaling pathways involved in immunosuppressive cells and briefly discuss how cancer cells evade immunosurveillance by manipulating antigen processing cells and related proteins. Secondly, the review will discuss how these pathways can be utilized for the implementation of precision medicine and deciphering drug resistance.
Collapse
|
18
|
Setlai BP, Hull R, Bida M, Durandt C, Mulaudzi TV, Chatziioannou A, Dlamini Z. Immunosuppressive Signaling Pathways as Targeted Cancer Therapies. Biomedicines 2022; 10:682. [PMID: 35327484 PMCID: PMC8945019 DOI: 10.3390/biomedicines10030682] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 12/23/2022] Open
Abstract
Immune response has been shown to play an important role in defining patient prognosis and response to cancer treatment. Tumor-induced immunosuppression encouraged the recent development of new chemotherapeutic agents that assists in the augmentation of immune responses. Molecular mechanisms that tumors use to evade immunosurveillance are attributed to their ability to alter antigen processing/presentation pathways and the tumor microenvironment. Cancer cells take advantage of normal molecular and immunoregulatory machinery to survive and thrive. Cancer cells constantly adjust their genetic makeup using several mechanisms such as nucleotide excision repair as well as microsatellite and chromosomal instability, thus giving rise to new variants with reduced immunogenicity and the ability to continue to grow without restrictions. This review will focus on the central molecular signaling pathways involved in immunosuppressive cells and briefly discuss how cancer cells evade immunosurveillance by manipulating antigen processing cells and related proteins. Secondly, the review will discuss how these pathways can be utilized for the implementation of precision medicine and deciphering drug resistance.
Collapse
Affiliation(s)
- Botle Precious Setlai
- Department of Surgery, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia 0007, South Africa;
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa;
| | - Meshack Bida
- Department of Anatomical Pathology, National Health Laboratory Service (NHLS), University of Pretoria, Hatfield 0028, South Africa;
| | - Chrisna Durandt
- Institute for Cellular and Molecular Medicine, Department of Immunology, SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| | - Thanyani Victor Mulaudzi
- Department of Surgery, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia 0007, South Africa;
| | - Aristotelis Chatziioannou
- Center of Systems Biology, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Str., 115 27 Athens, Greece;
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa;
| |
Collapse
|
19
|
Miki S, Suzuki JI, Takashima M, Ishida M, Kokubo H, Yoshizumi M. S-1-Propenylcysteine promotes IL-10-induced M2c macrophage polarization through prolonged activation of IL-10R/STAT3 signaling. Sci Rep 2021; 11:22469. [PMID: 34789834 PMCID: PMC8599840 DOI: 10.1038/s41598-021-01866-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/02/2021] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that may lead to the development of serious cardiovascular diseases. Aged garlic extract (AGE) has been reported to ameliorate atherosclerosis, although its mode of action remains unclear. We found that AGE increased the mRNA or protein levels of arginase1 (Arg1), interleukin-10 (IL-10), CD206 and hypoxia-inducible factor 2α (HIF2α) and decreased that of CD68, HIF1α and inducible nitric oxide synthase in the aorta and spleen of apolipoprotein E knockout mice. We also found that S-1-propenylcysteine (S1PC), a characteristic sulfur compound in AGE, increased the level of IL-10-induced Arg1 mRNA and the extent of M2c-like macrophage polarization in vitro. In addition, S1PC increased the population of M2c-like macrophages, resulting in suppressed the population of M1-like macrophages and decreased lipopolysaccharide-induced production of pro-inflammatory cytokines. These effects were accompanied by prolonged phosphorylation of the IL-10 receptor α (IL-10Rα) and signal transducer and activator of transcription 3 (STAT3) that inhibited the interaction between IL-10Rα and Src homology-2-containing inositol 5'-phosphatase 1 (SHIP1). In addition, administration of S1PC elevated the M2c/M1 macrophage ratio in senescence-accelerated mice. These findings suggest that S1PC may help improve atherosclerosis due to its anti-inflammatory effect to promote IL-10-induced M2c macrophage polarization.
Collapse
Affiliation(s)
- Satomi Miki
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima-shi, Hiroshima, 734-8551, Japan.
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., 1624 Shimokotachi, Koda-cho, Akitakata-shi, Hiroshima, 739-1195, Japan.
| | - Jun-Ichiro Suzuki
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., 1624 Shimokotachi, Koda-cho, Akitakata-shi, Hiroshima, 739-1195, Japan
| | - Miyuki Takashima
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., 1624 Shimokotachi, Koda-cho, Akitakata-shi, Hiroshima, 739-1195, Japan
| | - Mari Ishida
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima-shi, Hiroshima, 734-8551, Japan
| | - Hiroki Kokubo
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima-shi, Hiroshima, 734-8551, Japan
| | - Masao Yoshizumi
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima-shi, Hiroshima, 734-8551, Japan.
| |
Collapse
|
20
|
Ni D, Chai Z, Wang Y, Li M, Yu Z, Liu Y, Lu S, Zhang J. Along the allostery stream: Recent advances in computational methods for allosteric drug discovery. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2021. [DOI: 10.1002/wcms.1585] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Duan Ni
- College of Pharmacy Ningxia Medical University Yinchuan China
- The Charles Perkins Centre University of Sydney Sydney New South Wales Australia
| | - Zongtao Chai
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital Second Military Medical University Shanghai China
| | - Ying Wang
- State Key Laboratory of Oncogenes and Related Genes, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Mingyu Li
- State Key Laboratory of Oncogenes and Related Genes, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education Shanghai Jiao Tong University School of Medicine Shanghai China
| | | | - Yaqin Liu
- Medicinal Chemistry and Bioinformatics Center Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Shaoyong Lu
- College of Pharmacy Ningxia Medical University Yinchuan China
- State Key Laboratory of Oncogenes and Related Genes, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education Shanghai Jiao Tong University School of Medicine Shanghai China
- Medicinal Chemistry and Bioinformatics Center Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Jian Zhang
- College of Pharmacy Ningxia Medical University Yinchuan China
- State Key Laboratory of Oncogenes and Related Genes, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education Shanghai Jiao Tong University School of Medicine Shanghai China
- Medicinal Chemistry and Bioinformatics Center Shanghai Jiao Tong University School of Medicine Shanghai China
- School of Pharmaceutical Sciences Zhengzhou University Zhengzhou China
| |
Collapse
|
21
|
Islam H, Neudorf H, Mui AL, Little JP. Interpreting 'anti-inflammatory' cytokine responses to exercise: focus on interleukin-10. J Physiol 2021; 599:5163-5177. [PMID: 34647335 DOI: 10.1113/jp281356] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022] Open
Abstract
Circulating concentrations of canonically pro- and anti-inflammatory cytokines are commonly measured when evaluating the anti-inflammatory effects of exercise. An important caveat to interpreting systemic cytokine concentrations as evidence for the anti-inflammatory effects of exercise is the observed dissociation between circulating cytokine concentrations and cytokine function at the tissue/cellular level. The dichotomization of cytokines as pro- or anti-inflammatory also overlooks the context dependence of cytokine function, which can vary depending on the physiological state being studied, the cytokine's cellular source/target, and magnitude of cytokine responses. We re-evaluate our current understanding of anti-inflammatory cytokine responses to exercise by highlighting nuances surrounding the interpretation of altered systemic cytokine concentrations as evidence for changes in inflammatory processes occurring at the tissue/cellular level. We highlight the lesser known pro-inflammatory and immunostimulatory actions of the prototypical anti-inflammatory cytokine, interleukin (IL)-10, including the potentiation of interferon gamma production during endotoxaemia, CD8+ T cell activation in tumour bearing rodents and cancer patients in vivo, and CD8+ T lymphocyte and natural killer cell activation in vitro. IL-10's more well-established anti-inflammatory actions can also be blunted following exercise training and under chronic inflammatory states such as type 2 diabetes (T2D) independently of circulating IL-10 concentrations. The resistance to IL-10's anti-inflammatory action in T2D coincides with blunted STAT3 phosphorylation and can be restored with small-molecule activators of IL-10 signalling, highlighting potential therapeutic avenues for restoring IL-10 action. We posit that inferences based on altered circulating cytokine concentrations alone can miss important functional changes in cytokine action occurring at the tissue/cellular level.
Collapse
Affiliation(s)
- Hashim Islam
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Helena Neudorf
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Alice L Mui
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| |
Collapse
|
22
|
Islam H, Chamberlain TC, Mui AL, Little JP. Elevated Interleukin-10 Levels in COVID-19: Potentiation of Pro-Inflammatory Responses or Impaired Anti-Inflammatory Action? Front Immunol 2021; 12:677008. [PMID: 34234779 PMCID: PMC8255680 DOI: 10.3389/fimmu.2021.677008] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/08/2021] [Indexed: 12/12/2022] Open
Affiliation(s)
- Hashim Islam
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Thomas C Chamberlain
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Alice L Mui
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| |
Collapse
|
23
|
Whitfield H, Hemmings AM, Mills SJ, Baker K, White G, Rushworth S, Riley AM, Potter BVL, Brearley CA. Allosteric Site on SHIP2 Identified Through Fluorescent Ligand Screening and Crystallography: A Potential New Target for Intervention. J Med Chem 2021; 64:3813-3826. [PMID: 33724834 PMCID: PMC7610569 DOI: 10.1021/acs.jmedchem.0c01944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Src homology 2 domain-containing inositol phosphate phosphatase 2 (SHIP2) is one of the 10 human inositol phosphate 5-phosphatases. One of its physiological functions is dephosphorylation of phosphatidylinositol 3,4,5-trisphosphate, PtdIns(3,4,5)P3. It is therefore a therapeutic target for pathophysiologies dependent on PtdIns(3,4,5)P3 and PtdIns(3,4)P2. Therapeutic interventions are limited by the dearth of crystallographic data describing ligand/inhibitor binding. An active site-directed fluorescent probe facilitated screening of compound libraries for SHIP2 ligands. With two additional orthogonal assays, several ligands including galloflavin were identified as low micromolar Ki inhibitors. One ligand, an oxo-linked ethylene-bridged dimer of benzene 1,2,4-trisphosphate, was shown to be an uncompetitive inhibitor that binds to a regulatory site on the catalytic domain. We posit that binding of ligands to this site restrains L4 loop motions that are key to interdomain communications that accompany high catalytic activity with phosphoinositide substrate. This site may, therefore, be a future druggable target for medicinal chemistry.
Collapse
Affiliation(s)
- Hayley Whitfield
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| | - Andrew M Hemmings
- School of Chemistry, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| | - Stephen J Mills
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| | - Kendall Baker
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| | - Gaye White
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| | - Stuart Rushworth
- Department of Molecular Haematology; Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, U.K
| | - Andrew M Riley
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| | - Barry V L Potter
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| | - Charles A Brearley
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| |
Collapse
|
24
|
Targeting SHIP1 and SHIP2 in Cancer. Cancers (Basel) 2021; 13:cancers13040890. [PMID: 33672717 PMCID: PMC7924360 DOI: 10.3390/cancers13040890] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/12/2021] [Accepted: 02/13/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Phosphoinositol signaling pathways and their dysregulation have been shown to have a fundamental role in health and disease, respectively. The SH2-containing 5′ inositol phosphatases, SHIP1 and SHIP2, are regulators of the PI3K/AKT pathway that have crucial roles in cancer progression. This review aims to summarize the role of SHIP1 and SHIP2 in cancer signaling and the immune response to cancer, the discovery and use of SHIP inhibitors and agonists as possible cancer therapeutics. Abstract Membrane-anchored and soluble inositol phospholipid species are critical mediators of intracellular cell signaling cascades. Alterations in their normal production or degradation are implicated in the pathology of a number of disorders including cancer and pro-inflammatory conditions. The SH2-containing 5′ inositol phosphatases, SHIP1 and SHIP2, play a fundamental role in these processes by depleting PI(3,4,5)P3, but also by producing PI(3,4)P2 at the inner leaflet of the plasma membrane. With the intent of targeting SHIP1 or SHIP2 selectively, or both paralogs simultaneously, small molecule inhibitors and agonists have been developed and tested in vitro and in vivo over the last decade in various disease models. These studies have shown promising results in various pre-clinical models of disease including cancer and tumor immunotherapy. In this review the potential use of SHIP inhibitors in cancer is discussed with particular attention to the molecular structure, binding site and efficacy of these SHIP inhibitors.
Collapse
|