1
|
Parviainen E, Nurmenniemi S, Ravaioli S, Bravaccini S, Manninen A, Jukkola A, Selander K. Human papillomavirus E6 alters Toll-like receptor 9 transcripts and chemotherapy responses in breast cancer cells in vitro. Mol Biol Rep 2024; 52:43. [PMID: 39644451 PMCID: PMC11625066 DOI: 10.1007/s11033-024-10143-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Toll-like receptor 9 (TLR9) is a DNA recognizing receptor expressed also in several cancers. Decreased TLR9 expression is associated with poor prognosis in triple negative breast cancer (TNBC), but the role of TLR9 in breast cancer pathophysiology is currently unclear. Regulation of TLR9 expression in breast cancer is poorly understood. Human papillomavirus (HPV) infections suppress TLR9 expression in cervical cancers but the association between HPV and breast cancer has remained controversial. The aim of this study was to test if HPV16 can suppress TLR9 expression in breast cancer cells and affect cell behavior. METHODS AND RESULTS Human T-47D and MDA-MB-231 breast cancer cells were transduced with lentivirus encoding HPV16 E6 oncoprotein. The effects of E6 on TLR9 mRNA and protein expression, and cell proliferation, migration, invasion and sensitivity to chemotherapy were studied in vitro. Breast cancer tissue samples (n = 37) were analyzed for the presence of HPV DNA. E6 expression decreased TLR9 mRNA expression in MDA-MB-231 and T-47D cells in hypoxia. E6 expression altered breast cancer cell proliferation and made cells significantly less sensitive to the growth inhibitory effects of chemotherapeutic agents. HPV L1 gene was not detected in a small pilot cohort of clinical breast cancer specimens. CONCLUSION HPV16 may influence breast cancer cell TLR9 transcription and chemotherapy responses and could thereby affect breast cancer prognosis. These results suggest that HPV may have a previously unrecognized role in breast cancer pathophysiology and warrant further studies on the topic.
Collapse
Affiliation(s)
- Essi Parviainen
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Sini Nurmenniemi
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland.
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland.
| | - Sara Ravaioli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Bravaccini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
- Faculty of Medicine and Surgery, "Kore" University of Enna, Enna, Italy
| | - Aki Manninen
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Arja Jukkola
- Department of Oncology and Radiotherapy, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere Cancer Center, Tampere University, Tampere, Finland
| | - Katri Selander
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
- Department of Oncology and Radiation Therapy, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
2
|
Gangal H, Xie X, Huang Z, Cheng Y, Wang X, Lu J, Zhuang X, Essoh A, Huang Y, Chen R, Smith LN, Smith RJ, Wang J. Drug reinforcement impairs cognitive flexibility by inhibiting striatal cholinergic neurons. Nat Commun 2023; 14:3886. [PMID: 37391566 PMCID: PMC10313783 DOI: 10.1038/s41467-023-39623-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/20/2023] [Indexed: 07/02/2023] Open
Abstract
Addictive substance use impairs cognitive flexibility, with unclear underlying mechanisms. The reinforcement of substance use is mediated by the striatal direct-pathway medium spiny neurons (dMSNs) that project to the substantia nigra pars reticulata (SNr). Cognitive flexibility is mediated by striatal cholinergic interneurons (CINs), which receive extensive striatal inhibition. Here, we hypothesized that increased dMSN activity induced by substance use inhibits CINs, reducing cognitive flexibility. We found that cocaine administration in rodents caused long-lasting potentiation of local inhibitory dMSN-to-CIN transmission and decreased CIN firing in the dorsomedial striatum (DMS), a brain region critical for cognitive flexibility. Moreover, chemogenetic and time-locked optogenetic inhibition of DMS CINs suppressed flexibility of goal-directed behavior in instrumental reversal learning tasks. Notably, rabies-mediated tracing and physiological studies showed that SNr-projecting dMSNs, which mediate reinforcement, sent axonal collaterals to inhibit DMS CINs, which mediate flexibility. Our findings demonstrate that the local inhibitory dMSN-to-CIN circuit mediates the reinforcement-induced deficits in cognitive flexibility.
Collapse
Affiliation(s)
- Himanshu Gangal
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
- Institute for Neuroscience, Texas A&M University, College Station, TX, 77843, USA
| | - Xueyi Xie
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Zhenbo Huang
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Yifeng Cheng
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Xuehua Wang
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Jiayi Lu
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Xiaowen Zhuang
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Amanda Essoh
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
| | - Yufei Huang
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
- Institute for Neuroscience, Texas A&M University, College Station, TX, 77843, USA
| | - Ruifeng Chen
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, TX, 77843, USA
| | - Laura N Smith
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA
- Institute for Neuroscience, Texas A&M University, College Station, TX, 77843, USA
| | - Rachel J Smith
- Institute for Neuroscience, Texas A&M University, College Station, TX, 77843, USA
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Jun Wang
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, 77807, USA.
- Institute for Neuroscience, Texas A&M University, College Station, TX, 77843, USA.
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
3
|
Liao YH, Chan YH, Chen H, Yu AE, Sun LH, Yao WJ, Yu L. Stress while lacking of control induces ventral hippocampal autophagic flux hyperactivity and a depression-like behavior. Biomed J 2022; 45:896-906. [PMID: 34971825 PMCID: PMC9795357 DOI: 10.1016/j.bj.2021.12.008] [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: 07/25/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Stressed animals may perform depression-like behavior insomuch as stress-provoking blood-brain barrier (BBB) disruption, central immune activation, and autophagic flux changes. This study was undertaken to assess whether adult mice having (executive) vs. lacking (yoke) of behavioral control in otherwise equivalent stress magnitude condition, may display differences in their BBB integrity, ventral hippocampal (VH) interleukin-6 (IL-6) and autophagic flux level and VH-related depression-like behavior. To further understand the causative relation of enhanced autophagic flux and stress-primed depression-like behavior, we assessed the effects of bilateral intra-VH 3-methyladenine (3-MA), an autophagic flux inhibitor, infusion in stressed mice. METHODS Adult mice used had comparable genetic background and housing condition. Executive/yoke pairs of mice received a 10-day (1 h/day) footshock stressor regimen. Throughout the regimen, the ongoing footshock was terminated immediately contingent on the executive mouse', while irrelevant to the respective yoke mouse' voluntary behavior, or lasting for 7 s. Each dyad's cage-mate receiving no such regimen served as no stressor controls. RESULTS Yoke mice displayed disrupted BBB integrity (escalated Evans blue extravasation and decreased VH ZO-1, claudin-5 expression), increases in VH autophagic flux (increased LC3II/LC3I and decreased p62) and immobility duration in forced swimming test. Most of these indices remained unaltered in executive mice. Administration of 3-MA did not affect immobility duration in control mice, while prevented the increases in immobility duration in yoke mice. CONCLUSIONS (1) stress susceptibility may be determined by their differences in stress-coping results; (2) VH autophagic flux increase plays a permissive role in priming the stressed animals susceptible to exhibit depression-like behavior.
Collapse
Affiliation(s)
- Yi-Han Liao
- Department of Physiology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Ya-Hsuan Chan
- Department of Physiology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Hao Chen
- Department of Physiology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Anna E. Yu
- Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Li-Han Sun
- Institute of Basic Medical Sciences, and National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Wei-Jen Yao
- Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan,Corresponding author. Ditmanson Medical Foundation Chia-Yi Christian Hospital, 539, Zhongxiao Rd., East Dist., Chiayi 600566, Taiwan.
| | - Lung Yu
- Department of Physiology, National Cheng Kung University College of Medicine, Tainan, Taiwan,Institute of Basic Medical Sciences, and National Cheng Kung University College of Medicine, Tainan, Taiwan,Institute of Behavioral Medicine, National Cheng Kung University College of Medicine, Tainan, Taiwan,Corresponding author. Department of Physiology, National Cheng Kung University College of Medicine, Tainan, 70101 Taiwan.
| |
Collapse
|
4
|
Mohamed FEZ, Jalan R, Minogue S, Andreola F, Habtesion A, Hall A, Winstanley A, Damink SO, Malagó M, Davies N, Luong TV, Dhillon A, Mookerjee R, Dhar D, Al-Jehani RM. Inhibition of TLR7 and TLR9 Reduces Human Cholangiocarcinoma Cell Proliferation and Tumor Development. Dig Dis Sci 2022; 67:1806-1821. [PMID: 33939146 DOI: 10.1007/s10620-021-06973-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 03/25/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Toll-like receptors (TLRs) are key players in innate immunity and modulation of TLR signaling has been demonstrated to profoundly affect proliferation and growth in different types of cancer. However, the role of TLRs in human intrahepatic cholangiocarcinoma (ICC) pathogenesis remains largely unexplored. AIMS We set out to determine if TLRs play any role in ICCs which could potentially make them useful treatment targets. METHODS Tissue microarrays containing samples from 9 human ICCs and normal livers were examined immunohistochemically for TLR4, TLR7, and TLR9 expression. Proliferation of human ICC cell line HuCCT1 was measured by MTS assay following treatment with CpG-ODN (TLR9 agonist), imiquimod (TLR7 agonist), chloroquine (TLR7 and TLR9 inhibitor) and IRS-954 (TLR7 and TLR9 antagonist). The in vivo effects of CQ and IRS-954 on tumor development were also examined in a NOD-SCID mouse xenograft model of human ICC. RESULTS TLR4 was expressed in all normal human bile duct epithelium but absent in the majority (60%) of ICCs. TLR7 and TLR9 were expressed in 80% of human ICCs. However, TLR7 was absent in all cases of normal human bile duct epithelium and only one was TLR9 positive. HuCCT1 cell proliferation in vitro significantly increased following IMQ or CpG-ODN treatment (P < 0.03 and P < 0.002, respectively) but decreased with CQ (P < 0.02). In the mouse xenograft model there was significant reduction in size of tumors from CQ and IRS-954 treated mice compared to untreated controls. CONCLUSION TLR7 and TLR9 should be further explored for their potential as actionable targets in the treatment of ICC.
Collapse
Affiliation(s)
- Fatma El Zahraa Mohamed
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, UK.,Pathology Department, Minia University, El-Minia, Egypt
| | - Rajiv Jalan
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, UK
| | - Shane Minogue
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, UK
| | - Fausto Andreola
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, UK
| | - Abeba Habtesion
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, UK
| | - Andrew Hall
- UCL Institute for Liver and Digestive Health, Royal Free London NHS Foundation Trust, London, UK
| | - Alison Winstanley
- Department of Cellular Pathology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Steven Olde Damink
- Academic Department of Surgery and Interventional Sciences, Royal Free Hospital, London, UK
| | - Massimo Malagó
- Academic Department of Surgery and Interventional Sciences, Royal Free Hospital, London, UK
| | - Nathan Davies
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, UK
| | - Tu Vinh Luong
- Department of Cellular Pathology, Royal Free London NHS Foundation Trust, London, UK
| | - Amar Dhillon
- Department of Cellular Pathology, Royal Free London NHS Foundation Trust, London, UK
| | - Rajeshwar Mookerjee
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, UK
| | - Dipok Dhar
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, UK
| | - Rajai Munir Al-Jehani
- UCL Institute for Liver and Digestive Health, Royal Free London NHS Foundation Trust, London, UK.
| |
Collapse
|
5
|
Guan YH, Wang N, Deng ZW, Chen XG, Liu Y. Exploiting autophagy-regulative nanomaterials for activation of dendritic cells enables reinforced cancer immunotherapy. Biomaterials 2022; 282:121434. [DOI: 10.1016/j.biomaterials.2022.121434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/15/2022] [Accepted: 02/17/2022] [Indexed: 02/07/2023]
|
6
|
Costa TJ, Potje SR, Fraga-Silva TFC, da Silva-Neto JA, Barros PR, Rodrigues D, Machado MR, Martins RB, Santos-Eichler RA, Benatti MN, de Sá KSG, Almado CEL, Castro ÍA, Pontelli MC, Serra LL, Carneiro FS, Becari C, Louzada-Junior P, Oliveira RDR, Zamboni DS, Arruda E, Auxiliadora-Martins M, Giachini FRC, Bonato VLD, Zachara NE, Bomfim GF, Tostes RC. Mitochondrial DNA and TLR9 activation contribute to SARS-CoV-2-induced endothelial cell damage. Vascul Pharmacol 2021; 142:106946. [PMID: 34838735 PMCID: PMC8612754 DOI: 10.1016/j.vph.2021.106946] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/15/2021] [Indexed: 12/11/2022]
Abstract
Background and purpose Mitochondria play a central role in the host response to viral infection and immunity, being key to antiviral signaling and exacerbating inflammatory processes. Mitochondria and Toll-like receptor (TLR) have been suggested as potential targets in SARS-CoV-2 infection. However, the involvement of TLR9 in SARS-Cov-2-induced endothelial dysfunction and potential contribution to cardiovascular complications in COVID-19 have not been demonstrated. This study determined whether infection of endothelial cells by SARS-CoV-2 affects mitochondrial function and induces mitochondrial DNA (mtDNA) release. We also questioned whether TLR9 signaling mediates the inflammatory responses induced by SARS-CoV-2 in endothelial cells. Experimental approach Human umbilical vein endothelial cells (HUVECs) were infected by SARS-CoV-2 and immunofluorescence was used to confirm the infection. Mitochondrial function was analyzed by specific probes and mtDNA levels by real-time polymerase chain reaction (RT-PCR). Inflammatory markers were measured by ELISA, protein expression by western blot, intracellular calcium (Ca2+) by FLUOR-4, and vascular reactivity with a myography. Key results SARS-CoV-2 infected HUVECs, which express ACE2 and TMPRSS2 proteins, and promoted mitochondrial dysfunction, i.e. it increased mitochondria-derived superoxide anion, mitochondrial membrane potential, and mtDNA release, leading to activation of TLR9 and NF-kB, and release of cytokines. SARS-CoV-2 also decreased nitric oxide synthase (eNOS) expression and inhibited Ca2+ responses in endothelial cells. TLR9 blockade reduced SARS-CoV-2-induced IL-6 release and prevented decreased eNOS expression. mtDNA increased vascular reactivity to endothelin-1 (ET-1) in arteries from wild type, but not TLR9 knockout mice. These events were recapitulated in serum samples from COVID-19 patients, that exhibited increased levels of mtDNA compared to sex- and age-matched healthy subjects and patients with comorbidities. Conclusion and applications SARS-CoV-2 infection impairs mitochondrial function and activates TLR9 signaling in endothelial cells. TLR9 triggers inflammatory responses that lead to endothelial cell dysfunction, potentially contributing to the severity of symptoms in COVID-19. Targeting mitochondrial metabolic pathways may help to define novel therapeutic strategies for COVID-19.
Collapse
Affiliation(s)
- Tiago J Costa
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, USA.
| | - Simone R Potje
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil; Minas Gerais State University - UEMG, Brazil
| | - Thais F C Fraga-Silva
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Júlio A da Silva-Neto
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Paula R Barros
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Daniel Rodrigues
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Mirele R Machado
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Ronaldo B Martins
- Virology Research Center, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | | | - Maira N Benatti
- Department of Clinical Medicine, Division of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Keyla S G de Sá
- Department of Cell and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Carlos Eduardo L Almado
- Institute of Biological and Health Sciences, Federal University of Mato Grosso - UFMT, Brazil
| | - Ítalo A Castro
- Virology Research Center, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Marjorie C Pontelli
- Virology Research Center, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Leonardo La Serra
- Virology Research Center, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Fernando S Carneiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Christiane Becari
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Paulo Louzada-Junior
- Department of Clinical Medicine, Division of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Rene D R Oliveira
- Department of Clinical Medicine, Division of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Dario S Zamboni
- Department of Cell and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Eurico Arruda
- Virology Research Center, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Maria Auxiliadora-Martins
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Fernanda R C Giachini
- Institute of Biological and Health Sciences, Federal University of Mato Grosso - UFMT, Brazil
| | - Vânia L D Bonato
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil
| | - Natasha E Zachara
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, USA
| | - Gisele F Bomfim
- Institute of Health Sciences, Federal University of Mato Grosso - UFMT, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo - USP, Brazil.
| |
Collapse
|
7
|
Inhibition of Cathepsin K Alleviates Autophagy-Related Inflammation in Periodontitis-Aggravating Arthritis. Infect Immun 2020; 88:IAI.00498-20. [PMID: 32900814 DOI: 10.1128/iai.00498-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 02/05/2023] Open
Abstract
Rheumatoid arthritis (RA) and periodontitis share many epidemiological and pathological features, with emerging studies reporting a relationship between the two diseases. Recently, RA and periodontitis have been associated with autophagy. In the present study, we investigated the effects of cathepsin K (CtsK) inhibition on RA with periodontitis in a mouse model and its immunological function affecting autophagy. To topically inhibit CtsK periodontitis with arthritis in the animal model, adeno-associated virus (AAV) transfection was performed in periodontal and knee joint regions. Transfection of small interfering RNA (siRNA) was performed to inhibit CtsK in RAW264.7 cells. The effects of CtsK inhibition on the autophagy pathway were then evaluated in both in vivo and in vitro experiments. RA and periodontitis aggravated destruction and inflammation in their respective lesion areas. Inhibition of CtsK had multiple effects: (i) reduced destruction of alveolar bone and articular tissue, (ii) decreased macrophage numbers and inflammatory cytokine expression in the synovium, and (iii) alleviated expression of the autophagy-related transcription factor EB (TFEB) and microtubule-associated protein 1A/1B-light chain 3 (LC3) at the protein level in knee joints. Inhibition of CtsK in vitro reduced the expression of autophagy-related proteins and related inflammatory factors. Our data revealed that the inhibition of CtsK resisted the destruction of articular tissues and relieved inflammation from RA with periodontitis. Furthermore, CtsK was implicated as an imperative regulator of the autophagy pathway in RA and macrophages.
Collapse
|
8
|
Guo Z, Tang N, Liu FY, Yang Z, Ma SQ, An P, Wu HM, Fan D, Tang QZ. TLR9 deficiency alleviates doxorubicin-induced cardiotoxicity via the regulation of autophagy. J Cell Mol Med 2020; 24:10913-10923. [PMID: 33140921 PMCID: PMC7521247 DOI: 10.1111/jcmm.15719] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 07/05/2020] [Accepted: 07/09/2020] [Indexed: 12/19/2022] Open
Abstract
Doxorubicin is a commonly used anthracycline chemotherapeutic drug. Its application for treatment has been impeded by its cardiotoxicity as it is detrimental and fatal. DNA damage, cardiac inflammation, oxidative stress and cell death are the critical links in DOX-induced myocardial injury. Previous studies found that TLR9-related signalling pathways are associated with the inflammatory response of cardiac myocytes, mitochondrial dysfunction and cardiomyocyte death, but it remains unclear whether TLR9 could influence DOX-induced heart injury. Our current data imply that DOX-induced cardiotoxicity is ameliorated by TLR9 deficiency both in vivo and in vitro, manifested as improved cardiac function and reduced cardiomyocyte apoptosis and oxidative stress. Furthermore, the deletion of TLR9 rescued DOX-induced abnormal autophagy flux in vivo and in vitro. However, the inhibition of autophagy by 3-MA abolished the protective effects of TLR9 deletion on DOX-induced cardiotoxicity. Moreover, TLR9 ablation suppressed the activation of p38 MAPK during DOX administration and may promote autophagy via the TLR9-p38 MAPK signalling pathway. Our study suggests that the deletion of TLR9 exhibits a protective effect on doxorubicin-induced cardiotoxicity by enhancing p38-dependent autophagy. This finding could be used as a basis for the development of a prospective therapy against DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Nan Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Fang-Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Shu-Qing Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Peng An
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Hai-Ming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Di Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Ruiz A, Guzmán-Beltrán S, Carreto-Binaghi LE, Gonzalez Y, Juárez E. DNA from virulent M. tuberculosis induces TNF-α production and autophagy in M1 polarized macrophages. Microb Pathog 2019; 132:166-177. [PMID: 31054870 DOI: 10.1016/j.micpath.2019.04.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/15/2019] [Accepted: 04/29/2019] [Indexed: 12/23/2022]
Abstract
The macrophage innate immune response is outlined through recognition of the components of Mycobacterium tuberculosis. DNA of M. tuberculosis (MtbDNA) is recognized by macrophages, but the implications of this recognition are poorly characterized. Stimulation of murine macrophages with MtbDNA induces autophagy, a process that promotes elimination of intracellular pathogens. However, it remains unknown whether this or other phenomena also occur in human cells. In this work, we studied the innate response profiles of human macrophages after stimulation with DNA from virulent M. tuberculosis H37Rv. Human monocyte-derived macrophages were polarized into M1 and M2 phenotypes and stimulated with MtbDNA. The plasma membrane markers of the phenotype, production of TNF-α, and induction of autophagy were evaluated. Our results indicate that MtbDNA induced phenotypical changes, the significant production of TNF-α, and autophagy confirmed by the augmented expression of immunity related GTPase M (IRGM) and autophagy related ATG16L1 genes in M1 macrophages, whereas M2 macrophages exhibited limited responses. In addition, MtbDNA activation was TLR-9-dependent. Although TLR-9 expression was similar between M1 and M2 macrophages, only M1 macrophages were fully responsive to MtbDNA. In conclusion, MtbDNA recognition enhanced the antimicrobial mechanisms of M1 macrophages.
Collapse
Affiliation(s)
- Andy Ruiz
- Departamento de Investigación en Microbiología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, CDMX, 14080, Mexico; Posgrado en Ciencias Biológicas, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, 04510, Mexico
| | - Silvia Guzmán-Beltrán
- Departamento de Investigación en Microbiología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, CDMX, 14080, Mexico
| | - Laura E Carreto-Binaghi
- Departamento de Investigación en Microbiología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, CDMX, 14080, Mexico
| | - Yolanda Gonzalez
- Departamento de Investigación en Microbiología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, CDMX, 14080, Mexico
| | - Esmeralda Juárez
- Departamento de Investigación en Microbiología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, CDMX, 14080, Mexico.
| |
Collapse
|
10
|
Obergasteiger J, Frapporti G, Pramstaller PP, Hicks AA, Volta M. A new hypothesis for Parkinson's disease pathogenesis: GTPase-p38 MAPK signaling and autophagy as convergence points of etiology and genomics. Mol Neurodegener 2018; 13:40. [PMID: 30071902 PMCID: PMC6090926 DOI: 10.1186/s13024-018-0273-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 07/19/2018] [Indexed: 02/07/2023] Open
Abstract
The combination of genetics and genomics in Parkinson´s disease has recently begun to unveil molecular mechanisms possibly underlying disease onset and progression. In particular, catabolic processes such as autophagy have been increasingly gaining relevance as post-mortem evidence and experimental models suggested a participation in neurodegeneration and alpha-synuclein Lewy body pathology. In addition, familial Parkinson´s disease linked to LRRK2 and alpha-synuclein provided stronger correlation between etiology and alterations in autophagy. More detailed cellular pathways are proposed and genetic risk factors that associate with idiopathic Parkinson´s disease provide further clues in dissecting contributions of single players. Nevertheless, the fine-tuning of these processes remains elusive, as the initial stages of the pathways are not yet clarified.In this review, we collect literature evidence pointing to autophagy as the common, downstream target of Parkinsonian dysfunctions and augment current knowledge on the factors that direct the subsequent steps. Cell and molecular biology evidence indicate that p38 signaling underlies neurodegeneration and autoptic observations suggest a participation in neuropathology. Moreover, alpha-synuclein and LRRK2 also appear involved in the p38 pathway with additional roles in the regulation of GTPase signaling. Small GTPases are critical modulators of p38 activation and thus, their functional interaction with aSyn and LRRK2 could explain much of the detailed mechanics of autophagy in Parkinson´s disease.We propose a novel hypothesis for a more comprehensive working model where autophagy is controlled by upstream pathways, such as GTPase-p38, that have been so far underexplored in this context. In addition, etiological factors (LRRK2, alpha-synuclein) and risk loci might also combine in this common mechanism, providing a powerful experimental setting to dissect the cause of both familial and idiopathic disease.
Collapse
Affiliation(s)
- Julia Obergasteiger
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Giulia Frapporti
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Peter P. Pramstaller
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
- Department of Neurology, General Central Hospital, Via Böhler 5, 39100 Bolzano, Italy
- Department of Neurology, University of Lübeck, Ratzeburger Allee, 23538 Lübeck, Germany
| | - Andrew A. Hicks
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Mattia Volta
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| |
Collapse
|
11
|
Ye L, Feng Z, Doycheva D, Malaguit J, Dixon B, Xu N, Zhang JH, Tang J. CpG-ODN exerts a neuroprotective effect via the TLR9/pAMPK signaling pathway by activation of autophagy in a neonatal HIE rat model. Exp Neurol 2017; 301:70-80. [PMID: 29274721 DOI: 10.1016/j.expneurol.2017.12.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/28/2017] [Accepted: 12/19/2017] [Indexed: 12/16/2022]
Abstract
Hypoxic Ischemic Encephalopathy (HIE) is an injury caused to the brain due to prolonged lack of oxygen and blood supply which results in death or long-term disabilities. The main aim of this study was to investigate the role of Cytosine-phospho-guanine oligodeoxynucleotide (CpG-ODN) in autophagy after HIE. Ten-day old (P10) rat pups underwent right common carotid artery ligation followed by 2.5h of hypoxia as previously described by Rice-Vannucci. At 1h post HIE, rats were intranasally administered with recombinant CpG-ODN. Time-course expression levels of endogenous key proteins, TLR9, pAMPK/AMPK, LC3II/I, and LAMP1 involved in CpG-ODN's protective effects were measured using western blot. Short (48h) and long (4w) term neurobehavior studies were performed using righting reflex, negative geotaxis, water maze, foot fault and Rota rod tests. Brain samples were collected after long term for histological analysis. Furthermore, to elucidate the pathway via which CpG-ODN confers protection, TLR9 and AMPK inhibitors were used. Time course results showed that the expression of TLR9, pAMPK/AMPK, LC3II/I, LAMP1 increased after HIE. Neurobehavioral studies showed that HIE induced a significant delay in development and resulted in cognitive and motor function deficits. However, CpG-ODN ameliorated HIE-induced outcomes and improved long term neurological deficits. In addition, CpG-ODN increased expression of pAMPK/AMPK, p-ULK1/ULK1, P-AMBRA1/AMBRA1, LC3II/I and LAMP1 while inhibition of TLR9 and AMPK reversed those effects. In summary, CpG-ODN increased HIE-induced autophagy and improved short and long term neurobehavioral outcomes which may be mediated by the TLR9/pAMPK signaling pathway after HIE.
Collapse
Affiliation(s)
- Lan Ye
- The Medical Function Laboratory of Experimental Teaching Center of Basic Medicine, Guizhou Medical University, Guiyang 550004, China; Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States
| | - Zhanhui Feng
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States
| | - Desislava Doycheva
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States.
| | - Jay Malaguit
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States
| | - Brandon Dixon
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States.
| | - Ningbo Xu
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States
| | - John H Zhang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States; Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States; Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States
| | - Jiping Tang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92354, CA, United States.
| |
Collapse
|
12
|
Chandler MR, Keene KS, Tuomela JM, Forero-Torres A, Desmond R, Vuopala KS, Harris KW, Merner ND, Selander KS. Lower frequency of TLR9 variant associated with protection from breast cancer among African Americans. PLoS One 2017; 12:e0183832. [PMID: 28886076 PMCID: PMC5590816 DOI: 10.1371/journal.pone.0183832] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/11/2017] [Indexed: 12/11/2022] Open
Abstract
Introduction Toll-like receptor 9 (TLR9) is an innate immune system DNA-receptor that regulates tumor invasion and immunity in vitro. Low tumor TLR9 expression has been associated with poor survival in Caucasian patients with triple negative breast cancer (TNBC). African American (AA) patients with TNBC have worse prognosis than Caucasians but whether this is due to differences in tumor biology remains controversial. We studied the prognostic significance of tumor Toll like receptor-9 (TLR9) protein expression among African American (AA) triple negative breast cancer (TNBC) patients. Germline TLR9 variants in European Americans (EAs) and AAs were investigated, to determine their contribution to AA breast cancer risk. Methods TLR9 expression was studied with immunohistochemistry in archival tumors. Exome Variant Server and The Cancer Genome Atlas were used to determine the genetic variation in the general EA and AA populations, and AA breast cancer cases. Minor allele frequencies (MAFs) were compared between EAs (n = 4300), AAs (n = 2203), and/or AA breast cancer cases (n = 131). Results Thirty-two TLR9 variants had a statistically significant MAF difference between general EAs and AAs. Twenty-one of them affect a CpG site. Rs352140, a variant previously associated with protection from breast cancer, is more common in EAs than AAs (p = 2.20E-16). EAs had more synonymous alleles, while AAs had more rare coding alleles. Similar analyses comparing AA breast cancer cases with AA controls did not reveal any variant class differences; however, three previously unreported TLR9 variants were associated with late onset breast cancer. Although not statistically significant, rs352140 was observed less frequently in AA cases compared to controls. Tumor TLR9 protein expression was not associated with prognosis. Conclusions Tumor TLR9 expression is not associated with prognosis in AA TNBC. Significant differences were detected in TLR9 variant MAFs between EAs and AAs. They may affect TLR9 expression and function. Rs352140, which may protect from breast cancer, is 1.6 X more common among EAs. These findings call for a detailed analysis of the contribution of TLR9 to breast cancer pathophysiology and health disparities.
Collapse
Affiliation(s)
- Madison R. Chandler
- Harrison School of Pharmacy, Auburn University, Auburn, AL, United States of America
| | - Kimberly S. Keene
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Johanna M. Tuomela
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Andres Forero-Torres
- Department of Medicine, Division of Hematology & Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Renee Desmond
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Department of Medicine, Division of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Katri S. Vuopala
- Department of Pathology, Lapland Central Hospital, Rovaniemi, Finland
| | - Kevin W. Harris
- Department of Medicine, Division of Hematology & Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Birmingham Veterans Affairs Medical Center, Birmingham, AL, United States of America
| | - Nancy D. Merner
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
| | - Katri S. Selander
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States of America
- Department of Pathology, Lapland Central Hospital, Rovaniemi, Finland
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL, United States of America
- * E-mail:
| |
Collapse
|