1
|
Sciaccotta R, Gangemi S, Penna G, Giordano L, Pioggia G, Allegra A. Potential New Therapies "ROS-Based" in CLL: An Innovative Paradigm in the Induction of Tumor Cell Apoptosis. Antioxidants (Basel) 2024; 13:475. [PMID: 38671922 PMCID: PMC11047475 DOI: 10.3390/antiox13040475] [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: 03/18/2024] [Revised: 04/09/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Chronic lymphocytic leukemia, in spite of recent advancements, is still an incurable disease; the majority of patients eventually acquire resistance to treatment through relapses. In all subtypes of chronic lymphocytic leukemia, the disruption of normal B-cell homeostasis is thought to be mostly caused by the absence of apoptosis. Consequently, apoptosis induction is crucial to the management of this illness. Damaged biological components can accumulate as a result of the oxidation of intracellular lipids, proteins, and DNA by reactive oxygen species. It is possible that cancer cells are more susceptible to apoptosis because of their increased production of reactive oxygen species. An excess of reactive oxygen species can lead to oxidative stress, which can harm biological elements like DNA and trigger apoptotic pathways that cause planned cell death. In order to upset the balance of oxidative stress in cells, recent therapeutic treatments in chronic lymphocytic leukemia have focused on either producing reactive oxygen species or inhibiting it. Examples include targets created in the field of nanomedicine, natural extracts and nutraceuticals, tailored therapy using biomarkers, and metabolic targets. Current developments in the complex connection between apoptosis, particularly ferroptosis and its involvement in epigenomics and alterations, have created a new paradigm.
Collapse
Affiliation(s)
- Raffaele Sciaccotta
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.S.); (G.P.); (L.G.)
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Giuseppa Penna
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.S.); (G.P.); (L.G.)
| | - Laura Giordano
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.S.); (G.P.); (L.G.)
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Alessandro Allegra
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.S.); (G.P.); (L.G.)
| |
Collapse
|
2
|
Lopresti L, Capitani N, Tatangelo V, Tangredi C, Boncompagni G, Frezzato F, Visentin A, Marotta G, Ciofini S, Gozzetti A, Bocchia M, Trentin L, Baldari CT, Patrussi L. p66Shc deficiency in CLL cells enhances PD-L1 expression and suppresses immune synapse formation. Front Cell Dev Biol 2024; 12:1297116. [PMID: 38389706 PMCID: PMC10883382 DOI: 10.3389/fcell.2024.1297116] [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: 09/19/2023] [Accepted: 01/08/2024] [Indexed: 02/24/2024] Open
Abstract
Introduction: Escape from immunosurveillance is a hallmark of chronic lymphocytic leukemia (CLL) cells. In the protective niche of lymphoid organs, leukemic cells suppress the ability of T lymphocytes to form the immune synapse (IS), thereby hampering T-cell mediated anti-tumoral activities. By binding its cognate receptor PD-1 at the surface of T lymphocytes, the inhibitory ligand PD-L1, which is overexpressed in CLL cells, mediates the T-cell suppressive activities of CLL cells. However, the molecular mechanism underlying PD-L1 overexpression in CLL cells remains unknown. We have previously reported a defective expression of the pro-apoptotic and pro-oxidant adaptor p66Shc in CLL cells, which is causally related to an impairment in intracellular reactive oxygen species (ROS) production and to the activation of the ROS-sensitive transcription factor NF-κB. The fact that PD-L1 expression is regulated by NF-κB suggests a mechanistic relationship between p66Shc deficiency and PD-L1 overexpression in CLL cells. Methods: 62 treatment-naive CLL patients and 43 healthy donors were included in this study. PD-L1 and p66Shc expression was quantified in B cells by flow cytometry and qRT-PCR. IS architecture and local signaling was assessed by flow cytometry and confocal microscopy. CD8+ cell killing activity was assessed by flow cytometry. Results: Here we show that residual p66Shc expression in leukemic cells isolated both from CLL patients and from the CLL mouse model Eμ-TCL1 inversely correlated with PD-L1 expression. We also show that the PD-L1 increase prevented leukemic cells from forming ISs with T lymphocytes. Reconstitution of p66Shc, but not of a ROS-defective mutant, in both CLL cells and the CLL-derived cell line MEC-1, enhanced intracellular ROS and decreased PD-L1 expression. Similar results were obtained following treatment of CLL cells with H2O2 as exogenous source of ROS, that normalized PD-L1 expression and recovered IS formation. Discussion: Our data provide direct evidence that the p66Shc-deficiency-related ROS depletion in CLL cells concurs to enhance PD-L1 expression and provides a mechanistic basis for the suppression of T cell-mediated anti-tumoral functions in the immunosuppressive lymphoid niche.
Collapse
Affiliation(s)
| | - Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | | | | | - Federica Frezzato
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Andrea Visentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Giuseppe Marotta
- Stem Cell Transplant and Cellular Therapy Unit, University Hospital, Siena, Italy
| | - Sara Ciofini
- Department of Medical Science, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Alessandro Gozzetti
- Department of Medical Science, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Monica Bocchia
- Department of Medical Science, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Livio Trentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | | | - Laura Patrussi
- Department of Life Sciences, University of Siena, Siena, Italy
| |
Collapse
|
3
|
Ježek P, Jabůrek M, Holendová B, Engstová H, Dlasková A. Mitochondrial Cristae Morphology Reflecting Metabolism, Superoxide Formation, Redox Homeostasis, and Pathology. Antioxid Redox Signal 2023; 39:635-683. [PMID: 36793196 PMCID: PMC10615093 DOI: 10.1089/ars.2022.0173] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023]
Abstract
Significance: Mitochondrial (mt) reticulum network in the cell possesses amazing ultramorphology of parallel lamellar cristae, formed by the invaginated inner mitochondrial membrane. Its non-invaginated part, the inner boundary membrane (IBM) forms a cylindrical sandwich with the outer mitochondrial membrane (OMM). Crista membranes (CMs) meet IBM at crista junctions (CJs) of mt cristae organizing system (MICOS) complexes connected to OMM sorting and assembly machinery (SAM). Cristae dimensions, shape, and CJs have characteristic patterns for different metabolic regimes, physiological and pathological situations. Recent Advances: Cristae-shaping proteins were characterized, namely rows of ATP-synthase dimers forming the crista lamella edges, MICOS subunits, optic atrophy 1 (OPA1) isoforms and mitochondrial genome maintenance 1 (MGM1) filaments, prohibitins, and others. Detailed cristae ultramorphology changes were imaged by focused-ion beam/scanning electron microscopy. Dynamics of crista lamellae and mobile CJs were demonstrated by nanoscopy in living cells. With tBID-induced apoptosis a single entirely fused cristae reticulum was observed in a mitochondrial spheroid. Critical Issues: The mobility and composition of MICOS, OPA1, and ATP-synthase dimeric rows regulated by post-translational modifications might be exclusively responsible for cristae morphology changes, but ion fluxes across CM and resulting osmotic forces might be also involved. Inevitably, cristae ultramorphology should reflect also mitochondrial redox homeostasis, but details are unknown. Disordered cristae typically reflect higher superoxide formation. Future Directions: To link redox homeostasis to cristae ultramorphology and define markers, recent progress will help in uncovering mechanisms involved in proton-coupled electron transfer via the respiratory chain and in regulation of cristae architecture, leading to structural determination of superoxide formation sites and cristae ultramorphology changes in diseases. Antioxid. Redox Signal. 39, 635-683.
Collapse
Affiliation(s)
- Petr Ježek
- Department No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Martin Jabůrek
- Department No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Blanka Holendová
- Department No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Hana Engstová
- Department No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Andrea Dlasková
- Department No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
4
|
Chiu TH, Ku CW, Ho TJ, Tsai KL, Hsu WC, Chen YA, Ou HC, Chen HI. Schisanhenol Attenuates OxLDL-Induced Endothelial Dysfunction via an AMPK-Dependent Mechanism. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2023; 51:1459-1475. [PMID: 37518097 DOI: 10.1142/s0192415x23500660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Atherosclerotic cardiovascular diseases, commonly known as the formation of fibrofatty lesions in the artery wall, are the leading causes of death globally. Oxidized low-density lipoprotein (oxLDL) is one of the major components of atherosclerotic plaques. It is evident that dietary supplementation containing sources of antioxidants can prevent atherogenic diseases. Schisanhenol (SAL), a dibenzocyclooctene lignin, has been shown to attenuate oxLDL-induced apoptosis and the generation of reactive oxygen species (ROS) in endothelial cells. However, the underlying molecular mechanisms are still largely unknown. In this study, human umbilical vein endothelial cells (HUVECs) were pre-treated with SAL and oxLDL. Our results showed that adenosine monophosphate-activated protein kinase (AMPK) phosphorylation was enhanced in cells pre-treated with SAL in time-dependent and dose-dependent manners. Subsequently, oxLDL-induced AMPK dephosphorylation and protein kinase C (PKC) phosphorylation were significantly reversed in the presence of SAL. In addition, SAL treatment led to an inhibiting effect on the oxLDL-induced membrane assembly of NADPH oxidase subunits, and a similar effect was observed in ROS generation. This effect was further confirmed using knockdown AMPK with small interfering RNA (siRNA) and pharmaceutical reagents, such as the AMPK activator (AICAR), PKC inhibitor (Gö 6983), and ROS inhibitor (DPI). Furthermore, the oxLDL-induced intracellular calcium rise and the potential collapse of the mitochondrial membrane reduced the Bcl-2/Bax ratio, and released cytochrome c from the mitochondria, leading to the subsequent activation of caspase-3 in HUVECs, which were also markedly suppressed by SAL pretreatment. The results mentioned above may provide additional insights into the possible molecular mechanisms underlying the cardiovascular protective effects of SAL.
Collapse
Affiliation(s)
- Tsan-Hung Chiu
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Chang-Wen Ku
- Department of Chinese Medicine, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Kun-Ling Tsai
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Ching Hsu
- Department of Rehabilitation, Asia University Hospital, Taichung, Taiwan
| | - Yu-An Chen
- Department of Health and Leisure Management, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Hsiu-Chung Ou
- Department of Physical Therapy, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Hsiu-I Chen
- Department of Physical Therapy, College of Medical and Health Science, Asia University, Taichung, Taiwan
- Department of Physical Therapy, Hungkuang University, Taichung, Taiwan
| |
Collapse
|
5
|
Ding C, Yu Z, Sefik E, Zhou J, Kaffe E, Wang G, Li B, Flavell RA, Hu W, Ye Y, Li HB. A T reg-specific long noncoding RNA maintains immune-metabolic homeostasis in aging liver. NATURE AGING 2023; 3:813-828. [PMID: 37277640 DOI: 10.1038/s43587-023-00428-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 04/28/2023] [Indexed: 06/07/2023]
Abstract
Regulatory T (Treg) cells modulate several aging-related liver diseases. However, the molecular mechanisms regulating Treg function in this context are unknown. Here we identified a long noncoding RNA, Altre (aging liver Treg-expressed non-protein-coding RNA), which was specifically expressed in the nucleus of Treg cells and increased with aging. Treg-specific deletion of Altre did not affect Treg homeostasis and function in young mice but caused Treg metabolic dysfunction, inflammatory liver microenvironment, liver fibrosis and liver cancer in aged mice. Depletion of Altre reduced Treg mitochondrial integrity and respiratory capacity, and induced reactive oxygen species accumulation, thus increasing intrahepatic Treg apoptosis in aged mice. Moreover, lipidomic analysis identified a specific lipid species driving Treg aging and apoptosis in the aging liver microenvironment. Mechanistically, Altre interacts with Yin Yang 1 to orchestrate its occupation on chromatin, thereby regulating the expression of a group of mitochondrial genes, and maintaining optimal mitochondrial function and Treg fitness in the liver of aged mice. In conclusion, the Treg-specific nuclear long noncoding RNA Altre maintains the immune-metabolic homeostasis of the aged liver through Yin Yang 1-regulated optimal mitochondrial function and the Treg-sustained liver immune microenvironment. Thus, Altre is a potential therapeutic target for the treatment of liver diseases affecting older adults.
Collapse
Affiliation(s)
- Chenbo Ding
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhibin Yu
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Esen Sefik
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Jing Zhou
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Eleanna Kaffe
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Gaoyang Wang
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Weiguo Hu
- Department of Geriatrics, Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youqiong Ye
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua-Bing Li
- Medical Center on Aging, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Geriatrics, Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Chongqing International Institute for Immunology, Chongqing, China.
| |
Collapse
|
6
|
Tatangelo V, Boncompagni G, Capitani N, Lopresti L, Manganaro N, Frezzato F, Visentin A, Trentin L, Baldari CT, Patrussi L. p66Shc Deficiency in Chronic Lymphocytic Leukemia Promotes Chemokine Receptor Expression Through the ROS-Dependent Inhibition of NF-κB. Front Oncol 2022; 12:877495. [PMID: 35847884 PMCID: PMC9278989 DOI: 10.3389/fonc.2022.877495] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/01/2022] [Indexed: 12/02/2022] Open
Abstract
The microenvironment of lymphoid organs is central to the pathogenesis of chronic lymphocytic leukemia (CLL). Within it, tumor cells find a favourable niche to escape immunosurveillance and acquire pro-survival signals. We have previously reported that a CLL-associated defect in the expression of the pro-apoptotic and pro-oxidant adaptor p66Shc leads to enhanced homing to and accumulation of leukemic cells in the lymphoid microenvironment. The p66Shc deficiency-related impairment in intracellular reactive oxygen species (ROS) production in CLL cells is causally associated to the enhanced expression of the chemokine receptors CCR2, CXCR3 and CCR7, that promote leukemic cell homing to both lymphoid and non-lymphoid organs, suggesting the implication of a ROS-modulated transcription factor(s). Here we show that the activity of the ROS-responsive p65 subunit of the transcription factor NF-κB was hampered in the CLL-derived cell line MEC-1 expressing a NF-κB-luciferase reporter following treatment with H2O2. Similar results were obtained when intracellular ROS were generated by expression of p66Shc, but not of a ROS-defective mutant, in MEC-1 cells. NF-κB activation was associated with increased expression of the chemokine receptors CCR2, CXCR3 and CCR7. Reconstitution of p66Shc in CLL cells normalized intracellular ROS and hampered NF-κB activation, which led to a decrease in the expression of these homing receptors. Our data provide direct evidence that the p66Shc-deficiency-related ROS depletion in CLL cells concurs to NF-κB hyperactivation and homing receptor overexpression, providing a mechanistic basis for the enhanced ability of these cells to accumulate in the pro-survival lymphoid niche.
Collapse
Affiliation(s)
| | | | - Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | - Noemi Manganaro
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Federica Frezzato
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Andrea Visentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Livio Trentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | | | - Laura Patrussi
- Department of Life Sciences, University of Siena, Siena, Italy
| |
Collapse
|
7
|
Nicchi S, Giusti F, Carello S, Utrio Lanfaloni S, Tavarini S, Frigimelica E, Ferlenghi I, Rossi Paccani S, Merola M, Delany I, Scarlato V, Maione D, Brettoni C. Moraxella catarrhalis evades neutrophil oxidative stress responses providing a safer niche for nontypeable Haemophilus influenzae. iScience 2022; 25:103931. [PMID: 35265810 PMCID: PMC8899411 DOI: 10.1016/j.isci.2022.103931] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/20/2021] [Accepted: 02/10/2022] [Indexed: 11/25/2022] Open
Abstract
Moraxella catarrhalis and nontypeable Haemophilus influenzae (NTHi) are pathogenic bacteria frequently associated with exacerbation of chronic obstructive pulmonary disease (COPD), whose hallmark is inflammatory oxidative stress. Neutrophils produce reactive oxygen species (ROS) which can boost antimicrobial response by promoting neutrophil extracellular traps (NET) and autophagy. Here, we showed that M. catarrhalis induces less ROS and NET production in differentiated HL-60 cells compared to NTHi. It is also able to actively interfere with these responses in chemically activated cells in a phagocytosis and opsonin-independent and contact-dependent manner, possibly by engaging host immunosuppressive receptors. M. catarrhalis subverts the autophagic pathway of the phagocytic cells and survives intracellularly. It also promotes the survival of NTHi which is otherwise susceptible to the host antimicrobial arsenal. In-depth understanding of the immune evasion strategies exploited by these two human pathogens could suggest medical interventions to tackle COPD and potentially other diseases in which they co-exist. Mcat induces ROS and NET production to a lesser extent than NTHi in dHL-60 cells Mcat interferes with ROS-related responses in chemically-activated cells Mcat subverts the autophagic pathway surviving intracellularly while NTHi does not Intracellular survival of NTHi is enhanced by the co-infecting bacterium Mcat
Collapse
Affiliation(s)
- Sonia Nicchi
- GSK, Siena, 53100, Italy.,University of Bologna, Bologna, 40141, Italy
| | | | - Stefano Carello
- GSK, Siena, 53100, Italy.,University of Turin, Turin, 10100, Italy
| | | | | | | | | | | | - Marcello Merola
- GSK, Siena, 53100, Italy.,University of Naples Federico II, Naples, 80133, Italy
| | | | | | | | | |
Collapse
|
8
|
Liu R, Krüger K, Pilat C, Fan W, Xiao Y, Seimetz M, Ringseis R, Baumgart-Vogt E, Eder K, Weissmann N, Mooren FC. Excessive Accumulation of Intracellular Ca 2+ After Acute Exercise Potentiated Impairment of T-cell Function. Front Physiol 2021; 12:728625. [PMID: 34899372 PMCID: PMC8662941 DOI: 10.3389/fphys.2021.728625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 10/18/2021] [Indexed: 11/26/2022] Open
Abstract
Ca2+ is an important intracellular second messenger known to regulate several cellular functions. This research aimed to investigate the mechanisms of exercise-induced immunosuppression by measuring intracellular calcium levels, Ca2+-regulating gene expression, and agonist-evoked proliferation of murine splenic T lymphocytes. Mice were randomly assigned to the control, sedentary group (C), and three experimental groups, which performed a single bout of intensive and exhaustive treadmill exercise. Murine splenic lymphocytes were separated by density-gradient centrifugation immediately (E0), 3h (E3), and 24h after exercise (E24). Fura-2/AM was used to monitor cytoplasmic free Ca2+ concentration in living cells. The combined method of carboxyfluorescein diacetate succinimidyl ester (CFSE) labeling and flow cytometry was used for the detection of T cell proliferation. The transcriptional level of Ca2+-regulating genes was quantified by using qPCR. Both basal intracellular Ca2+ levels and agonist (ConA, OKT3, or thapsigargin)-induced Ca2+ transients were significantly elevated at E3 group (p<0.05 vs. control). However, mitogen-induced cell proliferation was significantly decreased at E3 group (p<0.05 vs. control). In parallel, the transcriptional level of plasma membrane Ca2+-ATPases (PMCA), sarco/endoplasmic reticulum Ca2+-ATPases (SERCA), TRPC1, and P2X7 was significantly downregulated, and the transcriptional level of IP3R2 and RyR2 was significantly upregulated in E3 (p<0.01 vs. control). In summary, this study demonstrated that acute exercise affected intracellular calcium homeostasis, most likely by enhancing transmembrane Ca2+ influx into cells and by reducing expression of Ca2+-ATPases such as PMCA and SERCA. However, altered Ca2+ signals were not transduced into an enhanced T cell proliferation suggesting other pathways to be responsible for the transient exercise-associated immunosuppression.
Collapse
Affiliation(s)
- Renyi Liu
- Department of Physical Education, China University of Geosciences (Wuhan), Wuhan, China.,Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Giessen, Germany
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Giessen, Germany
| | - Christian Pilat
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Giessen, Germany
| | - Wei Fan
- Institute for Anatomy and Cell Biology II, Justus-Liebig-University Giessen, Giessen, Germany
| | - Yu Xiao
- Institute for Anatomy and Cell Biology II, Justus-Liebig-University Giessen, Giessen, Germany
| | - Michael Seimetz
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Robert Ringseis
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology II, Justus-Liebig-University Giessen, Giessen, Germany
| | - Klaus Eder
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Frank Christoph Mooren
- Department of Rehabilitation Sciences, Faculty of Health, University of Witten/Herdecke, Witten, Germany
| |
Collapse
|
9
|
A novel class of oxazepine-based anti-cancer agents induces cell death in primary human CLL cells and efficiently reduces tumor growth in Eμ-TCL1 mice through the JNK/STAT4/p66Shc axis. Pharmacol Res 2021; 174:105965. [PMID: 34732370 DOI: 10.1016/j.phrs.2021.105965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/11/2021] [Accepted: 10/27/2021] [Indexed: 11/23/2022]
Abstract
Survival and expansion of malignant B cells in chronic lymphocytic leukemia (CLL) are highly dependent both on intrinsic defects in the apoptotic machinery and on the interactions with cells and soluble factors in the lymphoid microenvironment. The adaptor protein p66Shc is a negative regulator of antigen receptor signaling, chemotaxis and apoptosis whose loss in CLL B cells contributes to their extended survival and poor prognosis. Hence, the identification of compounds that restore p66Shc expression and function in malignant B cells may pave the way to a new therapeutic approach for CLL. Here we show that a novel oxazepine-based compound (OBC-1) restores p66Shc expression in primary human CLL cells by promoting JNK-dependent STAT4 activation without affecting normal B cells. Moreover, we demonstrate that the potent pro-apoptotic activity of OBC-1 in human leukemic cells directly correlates with p66Shc expression levels and is abrogated when p66Shc is genetically deleted. Preclinical testing of OBC-1 and the novel analogue OBC-2 in Eμ-TCL1 tumor-bearing mice resulted in a significantly longer overall survival and a reduction of the tumor burden in the spleen and peritoneum. Interestingly, OBCs promote leukemic cell mobilization from the spleen to the blood, which correlates with upregulation of sphingosine-1-phosphate receptor expression. In summary, our work identifies OBCs as a promising class of compounds that, by boosting p66Shc expression through the activation of the JNK/STAT4 pathway, display dual therapeutic effects for CLL intervention, namely the ability to mobilize cells from secondary lymphoid organs and a potent pro-apoptotic activity against circulating leukemic cells.
Collapse
|
10
|
Ku CW, Ho TJ, Huang CY, Chu PM, Ou HC, Hsieh PL. Cordycepin Attenuates Palmitic Acid-Induced Inflammation and Apoptosis of Vascular Endothelial Cells through Mediating PI3K/Akt/eNOS Signaling Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1703-1722. [PMID: 34488549 DOI: 10.1142/s0192415x21500804] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A well-known medicinal mushroom in the field of traditional Chinese medicine, Cordyceps sinensis, is a rare natural-occurring entomopathogenic fungus, and it typically grows at high altitudes on the plateau of the Himalayan. Previous studies indicated that cordycepin, the main bioactive chemical of Cordyceps sinensis, has very potent anticancer, anti-oxidant and anti-inflammatory activities. However, its protective effects against atherosclerotic changes in vascular endothelial cells have not been fully elucidated. In this study, we showed that pretreatment with cordycepin significantly attenuated palmitic acid (PA)-induced cytotoxicity, reactive oxygen species (ROS) generation, and inflammatory responses. We found that PA decreased phosphorylation of Akt, eNOS, and bioavailability of nitric oxide (NO), which in turn activated NF-[Formula: see text]B and the downstream inflammatory responses. All these detrimental events were markedly blocked by pretreatment with cordycepin. Moreover, cordycepin ameliorated destabilization of mitochondrial permeability, cytosolic calcium rises, and apoptotic features caused by PA. In addition, all these anti-inflammatory and anti-apoptosis effects of cordycepin were found to be inhibited by the PI3K and eNOS inhibitor, suggesting that its anti-atherosclerotic effects may partially be mediated by the PI3K/Akt/eNOS signaling pathway.
Collapse
Affiliation(s)
- Chang-Wen Ku
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan.,Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan.,Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan.,School of Post Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Department of Biotechnology, Asia University, Taichung, Taiwan.,Department of Medical Research, China Medical University, Taichung, Taiwan.,Holistic Education Center, Tzu Chi University of Science and Technology, Hualien, Taiwan
| | - Pei-Ming Chu
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Hsiu-Chung Ou
- Department of Physical Therapy, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Pei-Ling Hsieh
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
11
|
P66Shc: A Pleiotropic Regulator of B Cell Trafficking and a Gatekeeper in Chronic Lymphocytic Leukemia. Cancers (Basel) 2020; 12:cancers12041006. [PMID: 32325830 PMCID: PMC7226591 DOI: 10.3390/cancers12041006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/11/2020] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
Neoplastic B cells from chronic lymphocytic leukemia patients (CLL) have a profound deficiency in the expression of p66Shc, an adaptor protein with pro-apoptotic and pro-oxidant activities. This defect results in leukemic B cell resistance to apoptosis and additionally impinges on the balance between chemokine receptors that control B cell homing to secondary lymphoid organs and the sphingosine phosphate receptor S1PR1 that controls their egress therefrom, thereby favoring leukemic B cell accumulation in the pro-survival lymphoid niche. Ablation of the gene encoding p66Shc in the Eµ-TCL1 mouse model of human CLL enhances leukemogenesis and promotes leukemic cell invasiveness in both nodal and extranodal organs, providing in vivo evidence of the pathogenic role of the p66Shc defect in CLL pathogenesis. Here we present an overview of the functions of p66Shc in B lymphocytes, with a specific focus on the multiple mechanisms exploited by p66Shc to control B cell trafficking and the abnormalities in this process caused by p66Shc deficiency in CLL.
Collapse
|
12
|
Onnis A, Cassioli C, Finetti F, Baldari CT. Regulation of Selective B Cell Autophagy by the Pro-oxidant Adaptor p66SHC. Front Cell Dev Biol 2020; 8:193. [PMID: 32274384 PMCID: PMC7113388 DOI: 10.3389/fcell.2020.00193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/06/2020] [Indexed: 01/28/2023] Open
Abstract
p66SHC is a pro-oxidant member of the SHC family of protein adaptors that acts as a negative regulator of cell survival. In lymphocytes p66SHC exploits both its adaptor and its reactive oxygen species (ROS)-elevating function to antagonize mitogenic and survival signaling and promote apoptosis. As a result, p66SHC deficiency leads to the abnormal expansion of peripheral T and B cells and lupus-like autoimmunity. Additionally, a defect in p66SHC expression is a hallmark of B cell chronic lymphocytic leukemia, where it contributes to the accumulation of long-lived neoplastic cells. We have recently provided evidence that p66SHC exerts a further layer of control on B cell homeostasis by acting as a new mitochondrial LC3-II receptor to promote the autophagic demise of dysfunctional mitochondria. Here we discuss this finding in the context of the autophagic control of B cell homeostasis, development, and differentiation in health and disease.
Collapse
Affiliation(s)
- Anna Onnis
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Chiara Cassioli
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | | |
Collapse
|
13
|
Karunakaran U, Lee JE, Elumalai S, Moon JS, Won KC. Myricetin prevents thapsigargin-induced CDK5-P66Shc signalosome mediated pancreatic β-cell dysfunction. Free Radic Biol Med 2019; 141:59-66. [PMID: 31163256 DOI: 10.1016/j.freeradbiomed.2019.05.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 12/18/2022]
Abstract
Chronic endoplasmic reticulum (ER) stress has deleterious effects on pancreatic β-cell function and survival in type 2 diabetes (T2D). Cyclin-dependent kinase 5 (CDK5) plays a critical role in β-cell failure under diabetic milieu conditions. However, little information is available on CDK5's ability to impair the function of β-cells via a chemical ER stress inducer thapsigargin. Myricetin, a natural flavonoid, has therapeutic potential for the treatment of type 2 diabetes mellitus. Therefore, we examined the effect of CDK5 on thapsigargin-induced β-cell apoptosis, and explored the relationship between myricetin and CDK5. Exposure of beta cells with thapsigargin, induced a Src-mediated redox signaling (VAV2-Rac1-NOX) formation and CDK5 activation. Activated CDK5 induced antiapoptotic protein myeloid cell leukemia sequence 1 (Mcl-1) degradation which was associated with p66Shc serine 36 phosphorylation, causing beta cell apoptosis via mitochondrial dysfunction. Exposure of beta cells to myricetin resulted in an acute inhibition of Src-mediated redox signaling (VAV2-Rac1-NOX) formation and CDK5 activation. Myricetin inhibited CDK5 activation by directly binding to its ATP-binding pocket. Treatment with myricetin also enhanced the stability of Mcl-1 after thapsigargin treatment. Inhibition of CDK5 with myricetin or roscovitine, a CDK5 inhibitor attenuates thapsigargin induced p66Shc serine 36 phosphorylation and also reduced mitochondrial dysfunction by decreasing mitochondrial ROS and caspase-3 activation. In addition, myricetin was observed to enhance PDX-1 and insulin mRNA expression and potentiate glucose stimulated insulin secretion (GSIS). Taken together, these findings indicate that thapsigargin-induced early molecular events lead to CDK5-p66Shc signalosome contributes to thapsigargin-induced pancreatic β-cell dysfunction. Myricetin blocked thapsigargin induced CDK5-p66Shc signalosome formation and prevented pancreatic beta cell dysfunction. In this study, we demonstrated for the first time that thapsigargin initiated CDK5-p66Shc signalosome mediates the pancreatic beta cell dysfunction and myricetin protects the pancreatic beta cells through the inhibition of CDK5-p66Shc signalosome.
Collapse
Affiliation(s)
- Udayakumar Karunakaran
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Ji Eun Lee
- Department of Internal Medicine, CHA Gumi Medical Center, CHA University, Gumi, Republic of Korea
| | - Suma Elumalai
- Institute of Medical Science, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Jun Sung Moon
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea.
| | - Kyu Chang Won
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea; Institute of Medical Science, Yeungnam University College of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
14
|
Onnis A, Cianfanelli V, Cassioli C, Samardzic D, Pelicci PG, Cecconi F, Baldari CT. The pro-oxidant adaptor p66SHC promotes B cell mitophagy by disrupting mitochondrial integrity and recruiting LC3-II. Autophagy 2018; 14:2117-2138. [PMID: 30109811 DOI: 10.1080/15548627.2018.1505153] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Macroautophagy/autophagy has emerged as a central process in lymphocyte homeostasis, activation and differentiation. Based on our finding that the p66 isoform of SHC1 (p66SHC) pro-apoptotic ROS-elevating SHC family adaptor inhibits MTOR signaling in these cells, here we investigated the role of p66SHC in B-cell autophagy. We show that p66SHC disrupts mitochondrial function through its CYCS (cytochrome c, somatic) binding domain, thereby impairing ATP production, which results in AMPK activation and enhanced autophagic flux. While p66SHC binding to CYCS is sufficient for triggering apoptosis, p66SHC-mediated autophagy additionally depends on its ability to interact with membrane-associated LC3-II through a specific binding motif within its N terminus. Importantly, p66SHC also has an impact on mitochondria homeostasis by inducing mitochondrial depolarization, protein ubiquitination at the outer mitochondrial membrane, and local recruitment of active AMPK. These events initiate mitophagy, whose full execution relies on the role of p66SHC as an LC3-II receptor which brings phagophore membranes to mitochondria. Importantly, p66SHC also promotes hypoxia-induced mitophagy in B cells. Moreover, p66SHC deficiency enhances B cell differentiation to plasma cells, which is controlled by intracellular ROS levels and the hypoxic germinal center environment. The results identify mitochondrial p66SHC as a novel regulator of autophagy and mitophagy in B cells and implicate p66SHC-mediated coordination of autophagy and apoptosis in B cell survival and differentiation. Abbreviations: ACTB: actin beta; AMPK: AMP-activated protein kinase; ATP: adenosine triphosphate; ATG: autophagy-related; CYCS: cytochrome c, somatic; CLQ: chloroquine; COX: cyclooxygenase; CTR: control; GFP: green fluorescent protein; HIFIA/Hif alpha: hypoxia inducible factor 1 subunit alpha; IMS: intermembrane space; LIR: LC3 interacting region; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MTOR/mTOR: mechanistic target of rapamycin kinase; OA: oligomycin and antimycin A; OMM: outer mitochondrial membrane; PHB: prohibitin; PBS: phosphate-buffered saline; PINK1: PTEN induced putative kinase 1; RFP: red fluorescent protein; ROS: reactive oxygen species; SHC: src Homology 2 domain-containing transforming protein; TMRM: tetramethylrhodamine, methyl ester; TOMM: translocase of outer mitochondrial membrane; ULK1: unc-51 like autophagy activating kinase 1; WT: wild-type.
Collapse
Affiliation(s)
- Anna Onnis
- a Department of Life Sciences , University of Siena , Siena , Italy
| | - Valentina Cianfanelli
- b Cell Stress and Survival Unit , Danish Cancer Society Research Center , Copenhagen , Denmark
| | - Chiara Cassioli
- a Department of Life Sciences , University of Siena , Siena , Italy
| | - Dijana Samardzic
- c Venetian Institute of Molecular Medicine , University of Padova , Padova , Italy
| | - Pier Giuseppe Pelicci
- d Department of Experimental Oncology , European Institute of of Oncology , Milan , Italy
| | - Francesco Cecconi
- b Cell Stress and Survival Unit , Danish Cancer Society Research Center , Copenhagen , Denmark.,e Department of Biology , University of Rome Tor Vergata , Rome , Italy.,f Department of Pediatric Hematology and Oncology , Istituto di Ricovero e Cura a Carattere Scientifico Bambino Gesù Children's Hospital , Rome , Italy
| | - Cosima T Baldari
- a Department of Life Sciences , University of Siena , Siena , Italy
| |
Collapse
|
15
|
Mabruk ZA, Ahmed SBM, Thomas AC, Prigent SA. The role of the ShcD and RET interaction in neuroblastoma survival and migration. Biochem Biophys Rep 2018; 13:99-108. [PMID: 29556564 PMCID: PMC5857170 DOI: 10.1016/j.bbrep.2018.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 01/02/2018] [Accepted: 01/11/2018] [Indexed: 01/15/2023] Open
Abstract
Preliminary screening data showed that the ShcD adaptor protein associates with the proto-oncogene RET receptor tyrosine kinase. In the present study, we aimed to investigate the molecular interaction between ShcD and RET in human neuroblastoma cells and study the functional impact of this interaction. We were able to show that ShcD immunoprecipitated with RET from SK-N-AS neuroblastoma cell lysates upon GDNF treatment. This result was validated by ShcD-RET co-localization, which was visualized using a fluorescence microscope. ShcD-RET coexpression promoted ShcD and RET endosomal localization, resulting in unexpected inhibition of the downstream ERK and AKT pathways. Interestingly, ShcD-RET association reduced the viability and migration of SK-N-AS cells. Although ShcD was previously shown to trigger melanoma cell migration and tumorigenesis, our data showed an opposite role for ShcD in neuroblastoma SK-N-AS cells via its association with RET in GDNF-treated cells. In conclusion, ShcD acts as a switch molecule that promotes contrasting biological responses depending on the stimulus ad cell type. The melanoma associated Shc adaptor, ShcD, is found to interact with Ret oncogene receptor in SK-N-AS neuroblastoma cells. ShcD and Ret coexpression favoures their endosomal localization. ShcD-Ret association has suppressed ERK and AKT signalling. The functional consequence of ShcD and Ret interaction was shown to negatively affect cell survival and cellular migration in.
Collapse
Key Words
- ALK,, Anaplastic Lymphoma Kinase
- Akt,, Protein kinase B;
- CMV,, Cytomegalovirus
- DMEM,, Dulbecoo Modified Eagle's Medium;
- DNA,, Deoxyribonucleic Acid
- ECL,, Enhanced Chemiluminescence;
- EGF,, Epidermal Growth Factor;
- EGFR,, Epidermal Growth Factor Receptor;
- ERK,, Extracellular Signal–Regulated Kinases;
- Endosomes
- FBS,, Fetal Bovine Serum
- FGFR,, fibroblast growth factor receptors
- GDNF
- GDNF,, Glial Cell Line-Derived Neurotropic Factor;
- GFLs,, GDNF Family Ligands;
- GFP,, Green Fluorescent Protein
- GPCR,, G-Protein Coupled Receptor
- GRB2,, Growth Factor Receptor-Bound Protein 2;
- HGFR,, hepatocyte growth factor receptor;
- HRP,, Horseradish Peroxidase
- IGF,, Insulin Growth Factor;
- LB,, Luria-Bertani
- MAP,, Mitogen-Activated Protein;
- MAPK,, Mitogen-Activated Protein Kinases
- MuSK,, Muscle Specific Kinase
- NFDM,, Non-Fat Dry Milk
- Neuroblastoma
- PBS,, Phosphate-Buffered Saline
- PBST,, Phosphate-Buffered Saline Tween
- PDGF,, Platelet-Derived Growth Factor;
- PI3K,, Phosphoinositide 3-Kinase
- PMSF,, Phenylmethylsulfonyl Fluoride
- PVDF,, Polyvinylidene Fluoride
- RET
- RET,, Rearranged During Transfection
- RT,, Room Temperature;
- RTKs,, Receptor Tyrosine Kinase
- SDS-PAGE,, Sodium Dodecylsulphate Polyacrylamide Gel Electrophoresis
- ShcD
- ShcD,, Src Homology And Collagen D
- Src,, Proto-Oncogene Tyrosine-Protein Kinase Src
- TKRs,, Tyrosine Kinase Receptor;
- TrkA/B/C,, Tropomyosin-Related Kinase Receptor A/B/C
- hrs,, Hours
- mAb,, Monoclonal Antibody
- min,, Minute
- pAb,, Polyclonal Antibody
- pTyr,, Phospho-Tyrosine
- rpm,, revolution per minute;
Collapse
Affiliation(s)
- Zeanap A Mabruk
- Sharjah Institute for Medical Research and College of Medicine University of Sharjah, United Arab Emirates
| | - Samrein B M Ahmed
- Sharjah Institute for Medical Research and College of Medicine University of Sharjah, United Arab Emirates
| | - Asha Caroline Thomas
- Sharjah Institute for Medical Research and College of Medicine University of Sharjah, United Arab Emirates
| | - Sally A Prigent
- Department of Molecular and Cellular Biology, University of Leicester, UK
| |
Collapse
|
16
|
Stafford N, Wilson C, Oceandy D, Neyses L, Cartwright EJ. The Plasma Membrane Calcium ATPases and Their Role as Major New Players in Human Disease. Physiol Rev 2017; 97:1089-1125. [PMID: 28566538 DOI: 10.1152/physrev.00028.2016] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 01/20/2017] [Accepted: 01/23/2017] [Indexed: 02/07/2023] Open
Abstract
The Ca2+ extrusion function of the four mammalian isoforms of the plasma membrane calcium ATPases (PMCAs) is well established. There is also ever-increasing detail known of their roles in global and local Ca2+ homeostasis and intracellular Ca2+ signaling in a wide variety of cell types and tissues. It is becoming clear that the spatiotemporal patterns of expression of the PMCAs and the fact that their abundances and relative expression levels vary from cell type to cell type both reflect and impact on their specific functions in these cells. Over recent years it has become increasingly apparent that these genes have potentially significant roles in human health and disease, with PMCAs1-4 being associated with cardiovascular diseases, deafness, autism, ataxia, adenoma, and malarial resistance. This review will bring together evidence of the variety of tissue-specific functions of PMCAs and will highlight the roles these genes play in regulating normal physiological functions and the considerable impact the genes have on human disease.
Collapse
Affiliation(s)
- Nicholas Stafford
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Claire Wilson
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Ludwig Neyses
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
17
|
Abstract
Reactive oxygen species (ROS) are important signaling molecules that act through the oxidation of nucleic acids, proteins, and lipids. Several hallmarks of cancer, including uncontrolled proliferation, angiogenesis, and genomic instability, are promoted by the increased ROS levels commonly found in tumor cells. To counteract excessive ROS accumulation, oxidative stress, and death, cancer cells tightly regulate ROS levels by enhancing scavenging enzymes, which are dependent on the reducing cofactor nicotinamide adenine dinucleotide phosphate (NADPH). This review focuses on mitochondrial ROS homeostasis with a description of six pathways of NADPH production in mitochondria and a discussion of the possible strategies of pharmacological intervention to selectively eliminate cancer cells by increasing their ROS levels.
Collapse
Affiliation(s)
- Francesco Ciccarese
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Vincenzo Ciminale
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy.,Veneto Institute of Oncology - IRCCS, Padua, Italy
| |
Collapse
|
18
|
Plecitá-Hlavatá L, Ježek P. Integration of superoxide formation and cristae morphology for mitochondrial redox signaling. Int J Biochem Cell Biol 2016; 80:31-50. [PMID: 27640755 DOI: 10.1016/j.biocel.2016.09.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/09/2016] [Accepted: 09/12/2016] [Indexed: 12/16/2022]
Abstract
The mitochondrial network provides the central cell's energetic and regulatory unit, which besides ATP and metabolite production participates in cellular signaling through regulated reactive oxygen species (ROS) production and various protein/ion fluxes. The inner membrane forms extensive folds, called cristae, i.e. cavities enfolded from and situated perpendicularly to its inner boundary membrane portion, which encompasses an inner cylinder within the outer membrane tubule. Mitochondrial cristae ultramorphology reflects various metabolic, physiological or pathological states. Since the mitochondrion is typically a predominant superoxide source and generated ROS also serve for the creation of information redox signals, we review known relationships between ROS generation within the respiratory chain complexes of cristae and cristae morphology. Notably, it is emphasized that cristae shape is governed by ATP-synthase dimers, MICOS complexes, OPA1 isoforms and the umbrella of their regulation, and also dependent on local protonmotive force (electrical potential component) in cristae. Cristae are also affected by redox-sensitive kinases/phosphatases or p66SHC. ATP-synthase dimers decrease in the inflated intracristal space, diminishing pH and hypothetically having minimal superoxide formation. Matrix-released signaling superoxide/H2O2 is predominantly integrated along mitochondrial tubules, whereas the diffusion of intracristal signaling ROS species is controlled by crista junctions, the widening of which enables specific retrograde redox signaling such as during hypoxic cell adaptation. Other physiological cases of H2O2 release from the mitochondrion include the modulation of insulin release in pancreatic β-cells, enhancement of insulin signaling in peripheral tissues, signaling by T-cell receptors, retrograde signaling during the cell cycle and cell differentiation, specifically that of adipocytes.
Collapse
Affiliation(s)
- Lydie Plecitá-Hlavatá
- Department of Membrane Transport Biophysics, No.75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Petr Ježek
- Department of Membrane Transport Biophysics, No.75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| |
Collapse
|
19
|
Haller M, Khalid S, Kremser L, Fresser F, Furlan T, Hermann M, Guenther J, Drasche A, Leitges M, Giorgio M, Baier G, Lindner H, Troppmair J. Novel Insights into the PKCβ-dependent Regulation of the Oxidoreductase p66Shc. J Biol Chem 2016; 291:23557-23568. [PMID: 27624939 PMCID: PMC5095410 DOI: 10.1074/jbc.m116.752766] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Indexed: 12/12/2022] Open
Abstract
Dysfunctional mitochondria contribute to the development of many diseases and pathological conditions through the excessive production of reactive oxygen species (ROS), and, where studied, ablation of p66Shc (p66) was beneficial. p66 translocates to the mitochondria and oxidizes cytochrome c to yield H2O2, which in turn initiates cell death. PKCβ-mediated phosphorylation of serine 36 in p66 has been implicated as a key regulatory step preceding mitochondrial translocation, ROS production, and cell death, and PKCβ thus may provide a target for therapeutic intervention. We performed a reassessment of PKCβ regulation of the oxidoreductase activity of p66. Although our experiments did not substantiate Ser36 phosphorylation by PKCβ, they instead provided evidence for Ser139 and Ser213 as PKCβ phosphorylation sites regulating the pro-oxidant and pro-apoptotic function of p66. Mutation of another predicted PKCβ phosphorylation site also located in the phosphotyrosine binding domain, threonine 206, had no phenotype. Intriguingly, p66 with Thr206 and Ser213 mutated to glutamic acid showed a gain-of-function phenotype with significantly increased ROS production and cell death induction. Taken together, these data argue for a complex mechanism of PKCβ-dependent regulation of p66 activation involving Ser139 and a motif surrounding Ser213.
Collapse
Affiliation(s)
- Martina Haller
- From the Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery
| | - Sana Khalid
- From the Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery
| | - Leopold Kremser
- Division of Clinical Biochemistry, Protein Micro-Analysis Facility
| | - Friedrich Fresser
- Department for Pharmacology and Genetics, Division of Translational Cell Genetics, and
| | - Tobias Furlan
- From the Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery
| | - Martin Hermann
- Department for Anesthesiology and Intensive Care, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Julia Guenther
- From the Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery
| | - Astrid Drasche
- From the Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery
| | | | - Marco Giorgio
- the European Institute of Oncology, 20139 Milano, Italy
| | - Gottfried Baier
- Department for Pharmacology and Genetics, Division of Translational Cell Genetics, and
| | - Herbert Lindner
- Division of Clinical Biochemistry, Protein Micro-Analysis Facility
| | - Jakob Troppmair
- From the Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery,
| |
Collapse
|
20
|
Mlih M, Host L, Martin S, Niederhoffer N, Monassier L, Terrand J, Messaddeq N, Radke M, Gotthardt M, Bruban V, Kober F, Bernard M, Canet-Soulas E, Abt-Jijon F, Boucher P, Matz RL. The Src homology and collagen A (ShcA) adaptor protein is required for the spatial organization of the costamere/Z-disk network during heart development. J Biol Chem 2014; 290:2419-30. [PMID: 25488665 DOI: 10.1074/jbc.m114.597377] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Src homology and collagen A (ShcA) is an adaptor protein that binds to tyrosine kinase receptors. Its germ line deletion is embryonic lethal with abnormal cardiovascular system formation, and its role in cardiovascular development is unknown. To investigate its functional role in cardiovascular development in mice, ShcA was deleted in cardiomyocytes and vascular smooth muscle cells by crossing ShcA flox mice with SM22a-Cre transgenic mice. Conditional mutant mice developed signs of severe dilated cardiomyopathy, myocardial infarctions, and premature death. No evidence of a vascular contribution to the phenotype was observed. Histological analysis of the heart revealed aberrant sarcomeric Z-disk and M-band structures, and misalignments of T-tubules with Z-disks. We find that not only the ErbB3/Neuregulin signaling pathway but also the baroreceptor reflex response, which have been functionally associated, are altered in the mutant mice. We further demonstrate that ShcA interacts with Caveolin-1 and the costameric protein plasma membrane Ca(2+)/calmodulin-dependent ATPase (PMCA), and that its deletion leads to abnormal dystrophin signaling. Collectively, these results demonstrate that ShcA interacts with crucial proteins and pathways that link Z-disk and costamere.
Collapse
Affiliation(s)
- Mohamed Mlih
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Lionel Host
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Sophie Martin
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Nathalie Niederhoffer
- the Laboratory of Neurobiology and Cardiovascular Pharmacology Department, EA 7296, Federation of Translational Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Laurent Monassier
- the Laboratory of Neurobiology and Cardiovascular Pharmacology Department, EA 7296, Federation of Translational Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Jérôme Terrand
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Nadia Messaddeq
- the IGBMC, INSERM U964 CNRS UMR 7104, University of Strasbourg, 67401 Illkirch, France
| | - Michael Radke
- the Neuromuscular and Cardiovascular Cell Biology, Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany, the DZHK, German Centre for Cardiovascular Research, partner site, 13347 Berlin, Germany
| | - Michael Gotthardt
- the Neuromuscular and Cardiovascular Cell Biology, Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany, the DZHK, German Centre for Cardiovascular Research, partner site, 13347 Berlin, Germany
| | - Véronique Bruban
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Frank Kober
- the CRMBM, CNRS, UMR 7339, University of Aix-Marseille, 13385 Marseille, France, and
| | - Monique Bernard
- the CRMBM, CNRS, UMR 7339, University of Aix-Marseille, 13385 Marseille, France, and
| | - Emmanuelle Canet-Soulas
- the CREATIS-LRMN, CNRS, UMR 5220, U630 INSERM, 69621 Villeurbanne, Lyon-1 University, Lyon, France
| | | | - Philippe Boucher
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France,
| | - Rachel L Matz
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France,
| |
Collapse
|
21
|
Sánchez-Gómez FJ, Espinosa-Díez C, Dubey M, Dikshit M, Lamas S. S-glutathionylation: relevance in diabetes and potential role as a biomarker. Biol Chem 2014; 394:1263-80. [PMID: 24002664 DOI: 10.1515/hsz-2013-0150] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 06/10/2013] [Indexed: 02/06/2023]
Abstract
Glutathione is considered the main regulator of redox balance in the cellular milieu due to its capacity for detoxifying deleterious molecules. The oxidative stress induced as a result of a variety of stimuli promotes protein oxidation, usually at cysteine residues, leading to changes in their activity. Mild oxidative stress, which may take place in physiological conditions, induces the reversible oxidation of cysteines to sulfenic acid form, while pathological conditions are associated with higher rates of reactive oxygen species production, inducing the irreversible oxidation of cysteines. Among these, neurodegenerative disorders, cardiovascular diseases and diabetes have been proposed to be pathogenetically linked to this state. In diabetes-associated vascular complications, lower levels of glutathione and increased oxidative stress have been reported. S-glutathionylation has been proposed as a posttranslational modification able to protect proteins from over-oxidizing environments. S-glutathionylation has been identified in proteins involved in diabetic models both in vitro and in vivo. In all of them, S-glutathionylation represents a mechanism that regulates the response to diabetic conditions, and has been described to occur in erythrocytes and neutrophils from diabetic patients. However, additional studies are necessary to discern whether this modification represents a biomarker for the early onset of diabetic vascular complications.
Collapse
|
22
|
Mooren FC, Völker K, Klocke R, Nikol S, Waltenberger J, Krüger K. Exercise delays neutrophil apoptosis by a G-CSF-dependent mechanism. J Appl Physiol (1985) 2012; 113:1082-90. [PMID: 22858628 DOI: 10.1152/japplphysiol.00797.2012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The aim of the study was to determine whether exercise affects neutrophil apoptosis and to characterize the underlying mechanisms. Using annexin V labeling, neutrophil apoptosis was measured using flow cytometry after various bouts of exercise (marathon run, concentric/eccentric treadmill exercise, moderate/intensive resistance training) and in vitro conditions. Similarly, apoptosis-related markers as death receptors/ligands and mitochondrial membrane potential were detected. Furthermore, concentrations of intracellular free calcium and glutathione were measured using spectrofluorometry. After both marathon run and intensive laboratory exercise tests, neutrophil apoptosis was delayed. Furthermore, neutrophils mitochondrial membrane potential and death receptor/ligand expression were not affected by exercise. Apoptosis delay was accompanied under some exercise conditions by enhanced intracellular calcium transients and decreased glutathione levels. A delay of spontaneous apoptosis in vitro could be induced by incubation of neutrophils in postexercise serum. Heating of postexercise serum abolished the apoptosis delaying effect. In vitro stimulation of resting neutrophils with granulocyte-colony-stimulating factor (G-CSF) and C-reactive protein resulted in apoptosis delay too. Addition of anti-G-CSF antibody to postexercise serum was also effective in reversing its apoptosis-delaying effect. Exercise-induced mobilization of neutrophils is associated with a delay of apoptosis. This fundamental process seems to maintain exercise-induced neutrophilia and to contribute to the alerting and activation of the nonadaptive immune system known from other inflammatory conditions. An important extracellular trigger of apoptosis delay during exercise conditions seems to be G-CSF; intracellular processes may include calcium and redox signaling.
Collapse
Affiliation(s)
- Frank C Mooren
- Department of Sports Medicine, Institute of Sports Sciences, Justus-Liebig-University Giessen, Kugelberg 62, Giessen, Germany.
| | | | | | | | | | | |
Collapse
|
23
|
Kassab A, Piwowar A. Cell oxidant stress delivery and cell dysfunction onset in type 2 diabetes. Biochimie 2012; 94:1837-48. [PMID: 22333037 DOI: 10.1016/j.biochi.2012.01.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 01/25/2012] [Indexed: 01/18/2023]
Abstract
Most known pathways of diabetic complications involve oxidative stress. The mitochondria electron transport chain is a significant source of reactive oxygen species (ROS) in insulin secretory cells, insulin peripheral sensitive cells and endothelial cells. Elevated intracellular glucose level induces tricarboxylic acid cycle electron donor overproduction and mitochondrial proton gradient increase leading to an increase in electron transporter lifetime. Subsequently, the electrons leaked combine with respiratory oxygen (O(2)) resulting in superoxide anion ((•)O(2)(-)) production. Advanced glycation end products derive ROS via interaction with their receptors. Elevated diacylglycerol and ROS activate the protein kinase C pathway which, in turn, activates NADPH oxidases. A vicious circle of pathway derived ROS installs. Pathologic pathways induced ROS are activated and persistent though glycemia returns to normal due to hyperglycemia memory. Endothelial nitric oxide synthase may produce both superoxide anion ((•)O(2)(-)) and nitric oxide (NO) leading to peroxynitrite ((•)ONOO(-)) generation. Homocysteine is also implicated in oxidative stress pathogenesis. In this paper we have highlighted the pathologic mechanisms of ROS on atherosclerosis, renal dysfunction, retina dysfunction and nerve dysfunction in type 2 diabetes. Cell oxidant stress delivery have pivotal role in cell dysfunction onset and progression of angiopathies but an early introduction of good glycemic control may protect cells more efficiently than antioxidants.
Collapse
Affiliation(s)
- Asma Kassab
- Biochemistry Laboratory, CHU Farhat Hached, Sousse, Tunisia.
| | | |
Collapse
|
24
|
Borkowska A, Sielicka-Dudzin A, Herman-Antosiewicz A, Wozniak M, Fedeli D, Falcioni G, Antosiewicz J. Diallyl trisulfide-induced prostate cancer cell death is associated with Akt/PKB dephosphorylation mediated by P-p66shc. Eur J Nutr 2011; 51:817-25. [PMID: 22020565 PMCID: PMC3456917 DOI: 10.1007/s00394-011-0260-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 10/07/2011] [Indexed: 01/01/2023]
Abstract
Purpose P66Shc, an isoform of adaptor proteins, is known to mediate various signals including those leading to apoptosis or cell proliferation. Previously, we have shown that diallyl trisulfide (DATS)-induced prostate cancer cell death was mediated by increased ROS formation. In this study, we investigated the role of p66Shc protein and its serine 36 phosphorylation in DATS induced decrease in prostate cancer cell viability (PC-3). Methods PC-3 prostate cancer cells were used in this study. Stable cell lines expressing p66ShcS36A or an empty vector have been obtained. Cell viability, concentration of ROS, changes in P-p66Shc and P-Akt and DNA damage were determined. Results We observed that DATS treatment increased p66Shc phosphorylation at serine 36. Importantly, the phosphorylation was abolished by JNK inhibitor SP600125. Cells expressing plasmid-encoded variant of p66ShcS36A showed much higher resistance to DATS-induced cells death. In addition to that, we observed that DATS-induced ROS formation was completely abolished in cells expressing the p66ShcS36A variant. Interestingly, SP600125 proved to prevent DATS-induced Akt inactivation. In order to confirm that the observed effect is related to phosphorylation of p66Shc, we performed experiments on a stable cell line expressing p66ShcS36A. In such cells, DATS-induced Akt dephosphorylation was significantly reduced. On the other hand, hydrogen peroxide induced Akt activation in PC-3 cells, which was abrogated in cells expressing p66ShcS36A. Conclusions Our results uncover a novel signaling pathway with p66Shc being indispensable for DATS-induced inactivation of Akt due to hypophosphorylation.
Collapse
Affiliation(s)
- Andzelika Borkowska
- Department of Bioenergetics and Physiology of Exercise, Medical University of Gdańsk, Dębinki 1, 80-211, Gdańsk, Poland
| | | | | | | | | | | | | |
Collapse
|
25
|
Patrussi L, Giommoni N, Pellegrini M, Gamberucci A, Baldari CT. p66Shc-dependent apoptosis requires Lck and CamKII activity. Apoptosis 2011; 17:174-86. [DOI: 10.1007/s10495-011-0663-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
26
|
Numakawa T, Matsumoto T, Numakawa Y, Richards M, Yamawaki S, Kunugi H. Protective Action of Neurotrophic Factors and Estrogen against Oxidative Stress-Mediated Neurodegeneration. J Toxicol 2011; 2011:405194. [PMID: 21776259 PMCID: PMC3135156 DOI: 10.1155/2011/405194] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 02/28/2011] [Accepted: 03/29/2011] [Indexed: 01/01/2023] Open
Abstract
Oxidative stress is involved in the pathogenesis of neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. Low levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) are important for maintenance of neuronal function, though elevated levels lead to neuronal cell death. A complex series of events including excitotoxicity, Ca(2+) overload, and mitochondrial dysfunction contributes to oxidative stress-mediated neurodegeneration. As expected, many antioxidants like phytochemicals and vitamins are known to reduce oxidative toxicity. Additionally, growing evidence indicates that neurotrophic factors such as brain-derived neurotrophic factor (BDNF) and estrogens significantly prevent neuronal damage caused by oxidative stress. Here, we review and discuss recent studies addressing the protective mechanisms of neurotrophic factors and estrogen within this system.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
| | - Tomoya Matsumoto
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
- Department of Psychiatry and Neurosciences, Division of Frontier Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Yumiko Numakawa
- Peptide-prima Co., Ltd., 1-25-81, Nuyamazu, Kumamoto 861-2102, Japan
| | - Misty Richards
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
- The Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, NY 12208, USA
| | - Shigeto Yamawaki
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
- Department of Psychiatry and Neurosciences, Division of Frontier Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
- Core Research for Evolutional Science and Technology Program (CREST), Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
| |
Collapse
|
27
|
Ulivieri C, Fanigliulo D, Masi G, Savino MT, Gamberucci A, Pelicci PG, Baldari CT. p66Shc Is a Negative Regulator of FcεRI-Dependent Signaling in Mast Cells. THE JOURNAL OF IMMUNOLOGY 2011; 186:5095-106. [DOI: 10.4049/jimmunol.1001391] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
28
|
Fabbrocini G, Kisslinger A, Iannelli P, Vitale N, Procaccini C, Sparaneo G, Chieffi P, Ayala F, Mancini FP, Tramontano D. Resveratrol regulates p66Shc activation in HaCaT cells. Exp Dermatol 2011; 19:895-903. [PMID: 20626463 DOI: 10.1111/j.1600-0625.2010.01117.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skin is exposed to both endogenous and environmental oxidant agents, leading to the harmful generation of reactive oxygen species. Particular interest has been pointed on plant antioxidants, such as resveratrol, because of their wide-ranging biological activity and clinical potential. Resveratrol exerts antioxidant, metabolism-regulating and pro-apoptotic/anti-cancer effects on a variety of experimental models and has been suggested to protect skin from ultraviolet-induced photodamaging and photoaging. In parallel, also the biological significance of p66Shc, a member of the Src Homologue and Collagene homologue family with redox activity, is getting further attention. Because of the striking intersection among the activities of resveratrol with those of p66Shc, we investigated whether resveratrol would activate p66Shc in human immortalised keratinocytes (HaCaT cells), a well known and largely used model for skin keratinocytes. HaCaT cells were treated with resveratrol (10-150 μm) for different times. The effect of resveratrol on the proliferation of HaCaT cells and the activation of ERK1/2, AKT, and p66Shc was investigated by cell counting, fluorescence-activated cell sorting, and western blot analysis of total or immunoprecipitated cell extracts. In HaCaT cells, resveratrol induces dose- and time-dependent growth arrest, p66Shc-Ser36 phosphorylation, ERK1/2 phosphorylation and AKT dephosphorylation. Finally, we showed that resveratrol-induced p66Shc-Ser36 phosphorylation is dependent on ERK1/2 activation. Interestingly, these resveratrol-induced molecular effects were associated with reduced adhesion and reversible growth arrest rather than cell death pathways. This is the first evidence linking resveratrol with p66Shc and suggests that p66Shc may contribute to the effect of resveratrol on cell proliferation and function in the outermost layer of the skin.
Collapse
Affiliation(s)
- Gabriella Fabbrocini
- Department of Systematic Pathology, University of Naples Federico II, Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ulivieri C. Cell death: Insights into the ultrastructure of mitochondria. Tissue Cell 2010; 42:339-47. [DOI: 10.1016/j.tice.2010.10.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 10/18/2010] [Indexed: 02/08/2023]
|
30
|
Gertz M, Steegborn C. The Lifespan-regulator p66Shc in mitochondria: redox enzyme or redox sensor? Antioxid Redox Signal 2010; 13:1417-28. [PMID: 20214499 DOI: 10.1089/ars.2010.3147] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Mitochondria contribute to various diseases and aging phenotypes. Reactive oxygen species (ROS), mainly formed by the respiratory chain, were long thought to cause these effects by damaging proteins, DNA, and lipids. The emerging understanding that ROS act not only destructively but also as dedicated signaling molecules, and that aging processes are regulated by specific signaling networks has stimulated research on mitochondrial signaling systems and the regulation of mitochondrial ROS metabolism. p66Shc is a lifespan-regulating protein contributing to mitochondrial ROS metabolism and regulating the mitochondrial apoptosis pathway. It was found to participate in aging processes and has been implicated in several pathologies. Considerable progress has been made recently concerning the molecular function of p66Shc. It appears that p66Shc responds to a variety of proapoptotic stimuli by increasing ROS levels in the mitochondrial intermembrane space through an inherent ROS-producing activity, and that this ROS formation might trigger initiation of the mitochondrial apoptosis pathway. In this review, we will discuss the current knowledge on the molecular architecture of the p66Shc protein, its role in ROS metabolism and apoptosis regulation in the mitochondrial intermembrane space, the regulation of its mitochondrial transport, and the molecular mechanisms and interactions involved in these processes.
Collapse
Affiliation(s)
- Melanie Gertz
- Department of Physiological Chemistry, Ruhr-University Bochum, Germany
| | | |
Collapse
|
31
|
Capitani N, Lucherini OM, Baldari CT. Negative regulation of immunoreceptor signaling by protein adapters: Shc proteins join the club. FEBS Lett 2010; 584:4915-22. [DOI: 10.1016/j.febslet.2010.08.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 08/23/2010] [Accepted: 08/31/2010] [Indexed: 11/26/2022]
|
32
|
Finetti F, Savino MT, Baldari CT. Positive and negative regulation of antigen receptor signaling by the Shc family of protein adapters. Immunol Rev 2010; 232:115-34. [PMID: 19909360 DOI: 10.1111/j.1600-065x.2009.00826.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The Shc adapter family includes four members that are expressed as multiple isoforms and participate in signaling by a variety of cell-surface receptors. The biological relevance of Shc proteins as well as their variegated function, which relies on their highly conserved modular structure, is underscored by the distinct and dramatic phenotypic alterations resulting from deletion of individual Shc isoforms both in the mouse and in two model organisms, Drosophila melanogaster and Caenorhabditis elegans. The p52 isoform of ShcA couples antigen and cytokine receptors to Ras activation in both lymphoid and myeloid cells. However, the recognition of the spectrum of activities of p52ShcA in the immune system has been steadily expanding in recent years to other fundamental processes both at the cell and organism levels. Two other Shc family members, p66ShcA and p52ShcC/Rai, have been identified recently in T and B lymphocytes, where they antagonize survival and attenuate antigen receptor signaling. These developments reveal an unexpected and complex interplay of multiple Shc proteins in lymphocytes.
Collapse
Affiliation(s)
- Francesca Finetti
- Department of Evolutionary Biology, University of Siena, Siena, Italy
| | | | | |
Collapse
|
33
|
Essential role of the redox-sensitive kinase p66shc in determining energetic and oxidative status and cell fate in neuronal preconditioning. J Neurosci 2010; 30:5242-52. [PMID: 20392947 DOI: 10.1523/jneurosci.6366-09.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ischemic preconditioning is a phenomenon in which low-level stressful stimuli upregulate endogenous defensive programs, resulting in subsequent resistance to otherwise lethal injuries. We previously observed that signal transduction systems typically associated with neurodegeneration such as caspase activation are requisite events for the expression of tolerance and induction of HSP70. In this work, we sought to determine the extent and duration of oxidative and energetic dysfunction as well as the role of effector kinases on metabolic function in preconditioned cells. Using an in vitro neuronal culture model, we observed a robust increase in Raf and p66(Shc) activation within 1 h of preconditioning. Total ATP content decreased by 25% 3 h after preconditioning but returned to baseline by 24 h. Use of a free radical spin trap or p66(shc) inhibitor increased ATP content whereas a Raf inhibitor had no effect. Phosphorylated p66(shc) rapidly relocalized to the mitochondria and in the absence of activated p66(shc), autophagic processing increased. The constitutively expressed chaperone HSC70 relocalized to autophagosomes. Preconditioned cells experience significant total oxidative stress measured by F(2)-isoprostanes and neuronal stress evaluated by F(4)-neuroprostane measurement. Neuroprostane levels were enhanced in the presence of Shc inhibitors. Finally, we found that inhibiting either p66(shc) or Raf blocked neuroprotection afforded by preconditioning as well as upregulation of HSP70, suggesting both kinases are critical for preconditioning but function in fundamentally different ways. This is the first work to demonstrate the essential role of p66(shc) in mediating requisite mitochondrial and energetic compensation after preconditioning and suggests a mechanism by which protein and organelle damage mediated by ROS can increase HSP70.
Collapse
|
34
|
Rajendran M, Thomes P, Zhang L, Veeramani S, Lin MF. p66Shc--a longevity redox protein in human prostate cancer progression and metastasis : p66Shc in cancer progression and metastasis. Cancer Metastasis Rev 2010; 29:207-22. [PMID: 20111892 DOI: 10.1007/s10555-010-9213-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
p66Shc, a 66 kDa proto-oncogene Src homologous-collagen homologue (Shc) adaptor protein, is classically known in mediating receptor tyrosine kinase signaling and recently identified as a sensor to oxidative stress-induced apoptosis and as a longevity protein in mammals. The expression of p66Shc is decreased in mice and increased in human fibroblasts upon aging and in aging-related diseases, including prostate cancer. p66Shc protein level correlates with the proliferation of several carcinoma cells and can be regulated by steroid hormones. Recent advances point that p66Shc protein plays a role in mediating cross-talk between steroid hormones and redox signals by serving as a common convergence point in signaling pathways on cell proliferation and apoptosis. This article first reviews the unique function of p66Shc protein in regulating oxidative stress-induced apoptosis. Subsequently, we discuss its novel role in androgen-regulated prostate cancer cell proliferation and metastasis and the mechanism by which it mediates androgen action via the redox signaling pathway. The data together indicate that p66Shc might be a useful biomarker for the prognosis of prostate cancer and serve as an effective target for its cancer treatment.
Collapse
Affiliation(s)
- Mythilypriya Rajendran
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | | | | | | | | |
Collapse
|
35
|
Wang YN, Wu W, Chen HC, Fang H. Genistein protects against UVB-induced senescence-like characteristics in human dermal fibroblast by p66Shc down-regulation. J Dermatol Sci 2010; 58:19-27. [PMID: 20211546 DOI: 10.1016/j.jdermsci.2010.02.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2009] [Revised: 02/09/2010] [Accepted: 02/10/2010] [Indexed: 02/06/2023]
Abstract
BACKGROUND Genistein, as an active compound of dietary antioxidants, has shown considerable promise as an effective agent against aging process. However, the effect of genistein on skin photoaging and the associated mechanism remain unclear. OBJECTIVE To delineate the effect of genistein on UVB-induced senescence in human dermal fibroblasts (HDFs) with emphasis on the mechanism of oxidative pathway regulated by p66Shc involved in the events. METHODS HDFs were induced to premature senescence by repetitive subcytotoxic doses of UVB irradiation. Cellular apoptosis and DNA cell cycle were analyzed using flow cytometry. Intracellular levels of superoxide dismutase (SOD) and malondialdehyde (MDA) were detected by ELISA. Mutation levels of two large deletions of mitochondrial DNA, 4977bp and 3895bp deletion, were determined by quantitative PCR. Western blot was applied to detect the expression and activation of p66Shc (the 66-kilodalton isoform of the growth factor adapter Shc) and FKHRL1 (a forkhead protein that is intimately linked with intracellular oxidation). RESULTS Strong activity of senescence-associated beta-galactosidase (SA-beta-gal), high percent of cell apoptosis as well as cell cycle arrest in G0/G1 phase, and increased intracellular oxidative stress were observed in HDFs irradiated by UVB. Genistein exerted dramatically protective effects on HDFs in a dose-dependent manner. Elevated copy numbers of large deletions in mitochondrial DNA were also inhibited by genistein. Down-regulation of total and phosphorylated p66Shc on Ser36, as well as FKHRL1 and its phosphorylation on Thr32, were observed after genistein treatment. CONCLUSION The results indicate that genistein protects UVB-induced senescence-like characteristics in HDFs via maintenance of antioxidant enzyme activities and modulation of mitochondrial oxidative stress through down-regulation of a p66Shc-dependent signaling pathway, which may provide potential prevention against skin aging and even photoaging.
Collapse
Affiliation(s)
- Yi Na Wang
- Department of Dermatology, 1st Affiliated Hospital, Zhejiang University School of Medicine, 79# Qing Chun Road, Hangzhou 310003, China
| | | | | | | |
Collapse
|
36
|
Impaired expression of p66Shc, a novel regulator of B-cell survival, in chronic lymphocytic leukemia. Blood 2010; 115:3726-36. [PMID: 20061561 DOI: 10.1182/blood-2009-08-239244] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Intrinsic apoptosis defects underlie to a large extent the extended survival of malignant B cells in chronic lymphocytic leukemia (CLL). Here, we show that the Shc family adapter p66Shc uncouples the B-cell receptor (BCR) from the Erk- and Akt-dependent survival pathways, thereby enhancing B-cell apoptosis. p66Shc expression was found to be profoundly impaired in CLL B cells compared with normal peripheral B cells. Moreover, significant differences in p66Shc expression were observed in patients with favorable or unfavorable prognosis, based on the mutational status of IGHV genes, with the lowest expression in the unfavorable prognosis group. Analysis of the expression of genes implicated in apoptosis defects of CLL showed an alteration in the balance of proapoptotic and antiapoptotic members of the Bcl-2 family in patients with CLL. Reconstitution experiments in CLL B cells, together with data obtained on B cells from p66Shc(-/-) mice, showed that p66Shc expression correlates with a bias in the Bcl-2 family toward proapoptotic members. The data identify p66Shc as a novel regulator of B-cell apoptosis which attenuates BCR-dependent survival signals and modulates Bcl-2 family expression. They moreover provide evidence that the p66Shc expression defect in CLL B cells may be causal to the imbalance toward the antiapoptotic Bcl-2 family members in these cells.
Collapse
|
37
|
Kumar A, Hoffman TA, Dericco J, Naqvi A, Jain MK, Irani K. Transcriptional repression of Kruppel like factor-2 by the adaptor protein p66shc. FASEB J 2009; 23:4344-52. [PMID: 19696221 DOI: 10.1096/fj.09-138743] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The adaptor protein p66shc promotes cellular oxidative stress and apoptosis. Here, we demonstrate a novel mechanistic relationship between p66shc and the kruppel like factor-2 (KLF2) transcription factor and show that this relationship has biological relevance to p66shc-regulated cellular oxidant level, as well as KLF2-induced target gene expression. Genetic knockout of p66shc in mouse embryonic fibroblasts (MEFs) stimulates activity of the core KLF2 promoter and increases KLF2 mRNA and protein expression. Similarly, shRNA-induced knockdown of p66shc increases KLF2-promoter activity in HeLa cells. The increase in KLF2-promoter activity in p66shc-knockout MEFs is dependent on a myocyte enhancing factor-2A (MEF2A)-binding sequence in the core KLF2 promoter. Short-hairpin RNA-induced knockdown of p66shc in endothelial cells also stimulates KLF2 mRNA and protein expression, as well as expression of the endothelial KLF2 target gene thrombomodulin. MEF2A protein and mRNA are more abundant in p66shc-knockout MEFs, resulting in greater occupancy of the KLF2 promoter by MEF2A. In endothelial cells, the increase in KLF2 and thrombomodulin protein by shRNA-induced decrease in p66shc expression is partly abrogated by knockdown of MEF2A. Finally, knockdown of KLF2 abolishes the decrease in the cellular reactive oxygen species hydrogen peroxide observed with knockdown of p66shc, and KLF2 overexpression suppresses cellular hydrogen peroxide levels, independent of p66shc expression. These findings illustrate a novel mechanism by which p66shc promotes cellular oxidative stress, through suppression of MEF2A expression and consequent repression of KLF2 transcription.
Collapse
Affiliation(s)
- Ajay Kumar
- Cardiovascular Institute, University of Pittsburgh Medical Center, 623S Scaife Hall, 200 Lothrop St., Pittsburgh, PA 15213, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Graier WF, Malli R, Kostner GM. Mitochondrial protein phosphorylation: instigator or target of lipotoxicity? Trends Endocrinol Metab 2009; 20:186-93. [PMID: 19356948 PMCID: PMC4861235 DOI: 10.1016/j.tem.2009.01.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Revised: 01/21/2009] [Accepted: 01/21/2009] [Indexed: 01/28/2023]
Abstract
Lipotoxicity occurs as a consequence of chronic exposure of non-adipose tissue and cells to elevated concentrations of fatty acids, triglycerides and/or cholesterol. The contribution of mitochondria to lipotoxic cell dysfunction, damage and death is associated with elevated production of reactive oxygen species and initiation of apoptosis. Although there is a broad consensus on the involvement of these phenomena with lipotoxicity, the molecular mechanisms that initiate, mediate and trigger mitochondrial dysfunction in response to substrate overload remain unclear. Here, we focus on protein phosphorylation as an important phenomenon in lipotoxicity that harms mitochondria-related signal transduction and integration in cellular metabolism. Moreover, the degradation of mitochondria by mitophagy is discussed as an important landmark that leads to cellular apoptosis in lipotoxicity.
Collapse
Affiliation(s)
- Wolfgang F Graier
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University Graz, Graz, Austria.
| | | | | |
Collapse
|
39
|
Guo J, Gertsberg Z, Ozgen N, Steinberg SF. p66Shc links alpha1-adrenergic receptors to a reactive oxygen species-dependent AKT-FOXO3A phosphorylation pathway in cardiomyocytes. Circ Res 2009; 104:660-9. [PMID: 19168439 DOI: 10.1161/circresaha.108.186288] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
p66Shc is an adapter protein that is induced by hypertrophic stimuli and has been implicated as a major regulator of reactive oxygen species (ROS) production and cardiovascular oxidative stress responses. This study implicates p66Shc in an alpha(1)-adrenergtic receptor (alpha(1)-AR) pathway that requires the cooperative effects of protein kinase (PK)Cepsilon and PKCdelta and leads to AKT-FOXO3a phosphorylation in cardiomyocytes. alpha(1)-ARs promote p66Shc-YY(239/240) phosphorylation via a ROS-dependent mechanism that is localized to caveolae and requires epidermal growth factor receptor (EGFR) and PKCepsilon activity. alpha(1)-ARs also increase p66Shc-S(36) phosphorylation via an EGFR transactivation pathway involving PKCdelta. p66Shc links alpha(1)-ARs to an AKT signaling pathway that selectively phosphorylates/inactivates FOXO transcription factors and downregulates the ROS-scavenging protein manganese superoxide dismutase (MnSOD); the alpha(1)-AR-p66Shc-dependent pathway involving AKT does not regulate GSK3. Additional studies show that RNA interference-mediated downregulation of endogenous p66Shc leads to the derepression of FOXO3a-regulated genes such as MnSOD, p27Kip1, and BIM-1. p66Shc downregulation also increases proliferating cell nuclear antigen expression and induces cardiomyocyte hypertrophy, suggesting that p66Shc exerts an antihypertrophic action in neonatal cardiomyocytes. The novel alpha(1)-AR- and ROS-dependent pathway involving p66Shc identified in this study is likely to contribute to cardiomyocyte remodeling and the evolution of heart failure.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antibiotics, Antineoplastic/toxicity
- Apoptosis
- Cardiomegaly/metabolism
- Caveolae/metabolism
- Cell Enlargement
- Cells, Cultured
- Doxorubicin/toxicity
- ErbB Receptors/metabolism
- Forkhead Box Protein O3
- Forkhead Transcription Factors/metabolism
- Glycogen Synthase Kinase 3/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Norepinephrine/metabolism
- Oxidative Stress/drug effects
- Phosphorylation
- Protein Kinase C-delta/metabolism
- Protein Kinase C-epsilon/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- RNA Interference
- RNA, Small Interfering/metabolism
- Rats
- Rats, Wistar
- Reactive Oxygen Species/metabolism
- Receptors, Adrenergic, alpha-1/metabolism
- Shc Signaling Adaptor Proteins/genetics
- Shc Signaling Adaptor Proteins/metabolism
- Signal Transduction/drug effects
- Src Homology 2 Domain-Containing, Transforming Protein 1
- Time Factors
- Transduction, Genetic
Collapse
Affiliation(s)
- Jianfen Guo
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | | | | |
Collapse
|
40
|
Ulivieri C, Fanigliulo D, Benati D, Pasini FL, Baldari CT. Simvastatin impairs humoral and cell-mediated immunity in mice by inhibiting lymphocyte homing, T-cell activation and antigen cross-presentation. Eur J Immunol 2008; 38:2832-44. [PMID: 18958884 DOI: 10.1002/eji.200838278] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Statins block the activity of HMG-CoA reductase, which catalyses the production of mevalonate, an intermediate in cholesterol biosynthesis, which is also a precursor of isoprenoids. In addition to lowering circulating cholesterol, these drugs display anti-inflammatory and immunomodulatory activities in vitro; however, their effects on the development of adaptive immune responses in vivo, as well as the underlying mechanisms, are as yet largely unknown. Here we investigated the outcome of simvastatin treatment on a number of processes, which together orchestrate adaptive immunity to specific antigen. Simvastatin treatment resulted in a marked reduction of T and B cells in spleen, lymph nodes and peripheral blood in mice. This effect could be ascribed principally to an impairment of lymphocyte homing to secondary lymphoid organs. In addition, simvastatin was found to strongly inhibit T-cell responses to the MHCI restricted hen ovalbumin peptide antigen SIINFEKL and to impair ovalbumin uptake and cross-presentation by MHCI. Simvastatin also suppressed antibody responses to immunization with ovalbumin and delayed-type hypersensitivity to allergens. These activities could be largely accounted for by the simvastatin-dependent inhibition of HMG-CoA reductase. The data provide novel mechanistic insight into the activities of simvastatin in the highly complex context of the immune response.
Collapse
Affiliation(s)
- Cristina Ulivieri
- Department of Evolutionary Biology, University of Siena, Via Aldo Moro 2, Siena, Italy.
| | | | | | | | | |
Collapse
|
41
|
The proapoptotic and antimitogenic protein p66SHC acts as a negative regulator of lymphocyte activation and autoimmunity. Blood 2008; 111:5017-27. [DOI: 10.1182/blood-2007-12-130856] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The ShcA locus encodes 3 protein isoforms that differ in tissue specificity, subcellular localization, and function. Among these, p66Shc inhibits TCR coupling to the Ras/MAPK pathway and primes T cells to undergo apoptotic death. We have investigated the outcome of p66Shc deficiency on lymphocyte development and homeostasis. We show that p66Shc−/− mice develop an age-related lupus-like autoimmune disease characterized by spontaneous peripheral T- and B-cell activation and proliferation, autoantibody production, and immune complex deposition in kidney and skin, resulting in autoimmune glomerulonephritis and alopecia. p66Shc−/− lymphocytes display enhanced proliferation in response to antigen receptor engagement in vitro and more robust immune responses both to vaccination and to allergen sensitization in vivo. The data identify p66Shc as a negative regulator of lymphocyte activation and show that loss of this protein results in breaking of immunologic tolerance and development of systemic autoimmunity.
Collapse
|