1
|
Al Amin M, Bouhenni H, Zehravi M, Sweilam SH, Durgawale TP, Qureshi MS, Durgapal S, Haque MA, Vodeti R, Urs D, Shatu MM, Rab SO, Doukani K, Emran TB. Natural compounds and programmed necrosis: pioneering a new frontier in cancer treatments. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04050-w. [PMID: 40137962 DOI: 10.1007/s00210-025-04050-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025]
Abstract
Programmed necrosis, a controlled cell death method that bypasses resistance mechanisms that render apoptosis ineffective, is a potential cancer treatment target. Due to their diverse biological activities and low side effects, natural products are being explored as modulators of programmed necrosis pathways. This review highlights the potential of natural compounds to target cancer cells while preserving healthy tissues and their interaction with essential programmed necrosis mechanisms like ferroptosis and necroptosis. Recent developments have identified various types of programmable necrosis, including necroptosis, ferroptosis, pyroptosis, proptosis, mitochondrial permeability transition-driven necrosis, and oncosis. Natural compounds are increasingly being utilized as a primary source of anti-cancer medications, providing new cancer treatments. This review demonstrates the molecular mechanisms behind lipid peroxidation, mixed lineage kinase domain-like protein, and receptor-interacting protein kinases (RIPK1 and RIPK3) inducing cell death. Recent research has identified natural compounds like polyphenols, alkaloids, and terpenoids that can modulate pathways and benefit preclinical cancer models. The review underscores the potential of natural compounds in developing innovative cancer treatments by integrating pharmacology and cellular signaling knowledge. Integrating natural compound studies and programmed necrosis research presents a promising avenue for oncologists to overcome treatment resistance. Natural compounds have shown potential in developing programmed necrosis as a novel cancer treatment approach, enhancing therapeutic effectiveness and minimizing side effects through preclinical research, pharmacology, and molecular biology.
Collapse
Affiliation(s)
- Md Al Amin
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1216, Bangladesh.
| | - Hasna Bouhenni
- Laboratory of Agrobiotechnology and Nutrition in Semi-Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Saudi Arabia.
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo, 11829, Egypt
| | - Trupti Pratik Durgawale
- Department of Pharmaceutical Chemistry, Krishna Institute of Pharmacy Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, Maharashtra, India
| | - Mohammad Shamim Qureshi
- Department of Pharmacognosy & Phytochemistry, Anwarul Uloom College of Pharmacy, New Mallepally, Hyderabad, 500001, India
| | - Sumit Durgapal
- Department of Pharmaceutics, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Premnagar, Dehradun, Uttarakhand, 248007, India
| | - M Akiful Haque
- School of Pharmacy, Anurag University, Venkatapur, Hyderabad, Telangana , 500088, India
| | - Rajeshwar Vodeti
- Deportment of Pharmaceutics, School of Pharmacy, Anurag University, Hyderabad, India
| | - Deepadarshan Urs
- Inflammation Research Laboratory, Department of Studies & Research in Biochemistry, Mangalore University, Jnana Kaveri Post Graduate Centre, Kodagu, Karnataka, 571232, India
| | - Mst Maharunnasa Shatu
- Department of Botany, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Koula Doukani
- Laboratory of Agrobiotechnology and Nutrition in Semi-Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
- Laboratory of Animal Production Sciences and Techniques, Faculty of Nature and Life Sciences, University of Abdelhamid Ibn Badis, Mostaganem, Algeria
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1216, Bangladesh.
| |
Collapse
|
2
|
Li G, Liao C, Chen J, Wang Z, Zhu S, Lai J, Li Q, Chen Y, Wu D, Li J, Huang Y, Tian Y, Chen Y, Chen S. Targeting the MCP-GPX4/HMGB1 Axis for Effectively Triggering Immunogenic Ferroptosis in Pancreatic Ductal Adenocarcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308208. [PMID: 38593415 PMCID: PMC11151063 DOI: 10.1002/advs.202308208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 03/22/2024] [Indexed: 04/11/2024]
Abstract
Induction of ferroptosis can inhibit cancer cells in vitro, however, the role of ferroptosis in treatment in vivo is controversial. The immunosuppressive cells activated by the ferroptotic tumor cells can promote the growth of residual tumor cells, hindering the application of ferroptosis stimulation in tumor treatment. In this study, a new strategy is aimed to be identified for effectively triggering immunogenic ferroptosis in pancreatic ductal adenocarcinoma (PDAC) and simultaneously stimulating antitumor immune responses. Toward this, several molecular and biochemical experiments are performed using patient-derived organoid models and a KPC mouse model (LSL-KrasG12D /+, LSL-Trp53R172H/+, Pdx-1-Cre). It is observed that the inhibition of macrophage-capping protein (MCP) suppressed the ubiquitin fold modifier (UFM)ylation of pirin (PIR), a newly identified substrate of UFM1, thereby decreasing the transcription of GPX4, a marker of ferroptosis, and promoting the cytoplasmic transportation of HMGB1, a damage-associated molecular pattern. GPX4 deficiency triggered ferroptosis, and the pre-accumulated cytosolic HMGB1 is released rapidly. This altered release pattern of HMGB1 facilitated the pro-inflammatory M1-like polarization of macrophages. Thus, therapeutic inhibition of MCP yielded dual antitumor effects by stimulating ferroptosis and activating antitumor pro-inflammatory M1-like macrophages. The nanosystem developed for specifically silencing MCP is a promising tool for treating PDAC.
Collapse
Affiliation(s)
- Ge Li
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary SurgeryFujian Medical University Union HospitalFuzhou350001China
| | - Chengyu Liao
- Shengli Clinical Medical College of Fujian Medical UniversityFujian Medical UniversityFuzhou350001China
| | - Jiangzhi Chen
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary SurgeryFujian Medical University Union HospitalFuzhou350001China
| | - Zuwei Wang
- Shengli Clinical Medical College of Fujian Medical UniversityFujian Medical UniversityFuzhou350001China
- Department of Hepatopancreatobiliary SurgeryFujian Provincial HospitalFuzhou350001China
| | - Shuncang Zhu
- Shengli Clinical Medical College of Fujian Medical UniversityFujian Medical UniversityFuzhou350001China
| | - Jianlin Lai
- Shengli Clinical Medical College of Fujian Medical UniversityFujian Medical UniversityFuzhou350001China
- Department of Hepatopancreatobiliary SurgeryFujian Provincial HospitalFuzhou350001China
| | - Qiaowei Li
- Fujian Provincial Center for GeriatricsFuzhou350001China
- Fujian Key Laboratory of GeriatricsFuzhou350001China
| | - Yinhao Chen
- Shengli Clinical Medical College of Fujian Medical UniversityFujian Medical UniversityFuzhou350001China
| | - Dihan Wu
- Shengli Clinical Medical College of Fujian Medical UniversityFujian Medical UniversityFuzhou350001China
| | - Jianbo Li
- Shengli Clinical Medical College of Fujian Medical UniversityFujian Medical UniversityFuzhou350001China
| | - Yi Huang
- Shengli Clinical Medical College of Fujian Medical UniversityFujian Medical UniversityFuzhou350001China
- Center for Experimental Research in Clinical MedicineFujian Provincial HospitalFuzhou350001China
| | - Yifeng Tian
- Shengli Clinical Medical College of Fujian Medical UniversityFujian Medical UniversityFuzhou350001China
- Department of Hepatopancreatobiliary SurgeryFujian Provincial HospitalFuzhou350001China
| | - Yanling Chen
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary SurgeryFujian Medical University Union HospitalFuzhou350001China
| | - Shi Chen
- Shengli Clinical Medical College of Fujian Medical UniversityFujian Medical UniversityFuzhou350001China
- Department of Hepatopancreatobiliary SurgeryFujian Provincial HospitalFuzhou350001China
- Fujian Provincial Center for GeriatricsFuzhou350001China
- Fujian Key Laboratory of GeriatricsFuzhou350001China
| |
Collapse
|
3
|
Bielawski PB, Zhang I, Correa-Paz C, Campos F, Migliavacca M, Polo E, Del Pino P, Pelaz B, Vivien D, Maysinger D. Modulation of Abundance and Location of High-Mobility Group Box 1 in Human Microglia and Macrophages under Oxygen-Glucose Deprivation. ACS Pharmacol Transl Sci 2024; 7:680-692. [PMID: 38481701 PMCID: PMC10928888 DOI: 10.1021/acsptsci.3c00271] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/03/2024] [Accepted: 02/12/2024] [Indexed: 03/01/2025]
Abstract
While stroke represents one of the main causes of death worldwide, available effective drug treatment options remain limited to classic thrombolysis with recombinant tissue plasminogen activator (rtPA) for arterial-clot occlusion. Following stroke, multiple pathways become engaged in producing a vicious proinflammatory cycle through the release of damage-associated molecular patterns (DAMPs) such as high-mobility group box 1 (HMGB1) and heat shock protein 70 kDa (HSP72). HMGB1, in particular, can activate proinflammatory cytokine production when acetylated (AcHMGB1), a form that prefers cytosolic localization and extracellular release. This study aimed at determining how HMGB1 and HSP72 are modulated and affected following treatment with the anti-inflammatory compound resveratrol and novel platelet membrane-derived nanocarriers loaded with rtPA (CSM@rtPA) recently developed by our group for ischemic artery recanalization. Under ischemic conditions of oxygen-glucose deprivation (OGD), nuclear abundance of HMGB1 and AcHMGB1 in microglia and macrophages decreased, whereas treatment with CSM@rtPA did not alter nuclear or cytosolic abundance. Resveratrol treatment markedly increased the cytosolic abundance of HSP72 in microglia. Using proximity ligation assays, we determined that HSP72 interacted with HMGB1 and with acetylated HMGB1. The interaction was differentially affected under the OGD conditions. Resveratrol treatment under the OGD further decreased HSP72-HMGB1 interactions, whereas, in contrast, treatment increased HSP72-AcHMGB1 interactions in microglia. This study points out a salient molecular interaction suited for a two-pronged nanotherapeutic intervention in stroke: enhancement of rtPA's thrombolytic activity and modulation of cytosolic interactions between HMGB1 and HSP72 by resveratrol.
Collapse
Affiliation(s)
- Patrick-Brian Bielawski
- Department
of Pharmacology and Therapeutics, McGill
University, Montreal, QC H3G 1Y6, Canada
| | - Issan Zhang
- Department
of Pharmacology and Therapeutics, McGill
University, Montreal, QC H3G 1Y6, Canada
| | - Clara Correa-Paz
- Translational
Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory
(LINC), Health Research Institute of Santiago
de Compostela (IDIS), Santiago
de Compostela 15706, Spain
| | - Francisco Campos
- Translational
Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory
(LINC), Health Research Institute of Santiago
de Compostela (IDIS), Santiago
de Compostela 15706, Spain
| | - Martina Migliavacca
- Center
for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela (USC), Santiago de Compostela 15782, Spain
| | - Ester Polo
- Center
for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela (USC), Santiago de Compostela 15782, Spain
| | - Pablo Del Pino
- Center
for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela (USC), Santiago de Compostela 15782, Spain
| | - Beatriz Pelaz
- Center
for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela (USC), Santiago de Compostela 15782, Spain
| | - Denis Vivien
- UNICAEN,
INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders
(PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, 14000 Caen, France
- Department
of Clinical Research, Caen Normandie University
Hospital, 14000 Caen, France
| | - Dusica Maysinger
- Department
of Pharmacology and Therapeutics, McGill
University, Montreal, QC H3G 1Y6, Canada
| |
Collapse
|
4
|
Joma N, Zhang I, Righetto GL, McKay L, Gran ER, Kakkar A, Maysinger D. Flavonoids Regulate Redox-Responsive Transcription Factors in Glioblastoma and Microglia. Cells 2023; 12:2821. [PMID: 38132142 PMCID: PMC10871111 DOI: 10.3390/cells12242821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/29/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
The tumor microenvironment (TME) has emerged as a valuable therapeutic target in glioblastoma (GBM), as it promotes tumorigenesis via an increased production of reactive oxygen species (ROS). Immune cells such as microglia accumulate near the tumor and its hypoxic core, fostering tumor proliferation and angiogenesis. In this study, we explored the therapeutic potential of natural polyphenols with antioxidant and anti-inflammatory properties. Notably, flavonoids, including fisetin and quercetin, can protect non-cancerous cells while eliminating transformed cells (2D cultures and 3D tumoroids). We tested the hypothesis that fisetin and quercetin are modulators of redox-responsive transcription factors, for which subcellular location plays a critical role. To investigate the sites of interaction between natural compounds and stress-responsive transcription factors, we combined molecular docking with experimental methods employing proximity ligation assays. Our findings reveal that fisetin decreased cytosolic acetylated high mobility group box 1 (acHMGB1) and increased transcription factor EB (TFEB) abundance in microglia but not in GBM. Moreover, our results suggest that the most powerful modulator of the Nrf2-KEAP1 complex is fisetin. This finding is in line with molecular modeling and calculated binding properties between fisetin and Nrf2-KEAP1, which indicated more sites of interactions and stronger binding affinities than quercetin.
Collapse
Affiliation(s)
- Natali Joma
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada; (N.J.); (I.Z.); (G.L.R.); (E.R.G.)
| | - Issan Zhang
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada; (N.J.); (I.Z.); (G.L.R.); (E.R.G.)
| | - Germanna L. Righetto
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada; (N.J.); (I.Z.); (G.L.R.); (E.R.G.)
- Structural Genomics Consortium, University of Toronto, 101 College St, Toronto, ON M5G 1L7, Canada
| | - Laura McKay
- Department of Chemistry, McGill University, 801 Sherbrooke St W, Montreal, QC H3A 0B8, Canada; (L.M.); (A.K.)
| | - Evan Rizzel Gran
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada; (N.J.); (I.Z.); (G.L.R.); (E.R.G.)
| | - Ashok Kakkar
- Department of Chemistry, McGill University, 801 Sherbrooke St W, Montreal, QC H3A 0B8, Canada; (L.M.); (A.K.)
| | - Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, QC H3G 1Y6, Canada; (N.J.); (I.Z.); (G.L.R.); (E.R.G.)
| |
Collapse
|
5
|
Shao S, Xiao L, Jia M, Zhang C, Zhao G, Yao R, Wang X, Gao L. Never in mitosis gene A-related kinase-6 deficiency deteriorates diabetic cardiomyopathy via regulating heat shock protein 72. J Mol Med (Berl) 2023; 101:419-430. [PMID: 36867206 DOI: 10.1007/s00109-023-02295-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 03/04/2023]
Abstract
NIMA (never in mitosis, gene A)-related kinase-6 (NEK6), a cell cycle regulatory gene, was found to regulate cardiac hypertrophy. However, its role in diabetes-induced cardiomyopathy has not been fully elucidated. This research was designed to illustrate the effect of NEK6 involved in diabetic cardiomyopathy. Here we used a streptozotocin (STZ)-induced mice diabetic cardiomyopathy model and NEK6 knockout mice to explore the role and mechanism of NEK6 in diabetic-induced cardiomyopathy. NEK6 knockout mice and wild-type littermates were subjected to STZ injection (50 mg/kg/day for 5 days) to induce a diabetic cardiomyopathy model. As a result, 4 months after final STZ injection, DCM mice revealed cardiac hypertrophy, fibrosis, and systolic and diastolic dysfunction. NEK6 deficiency causes deteriorated cardiac hypertrophy, fibrosis, and cardiac dysfunction. Furthermore, we observed inflammation and oxidative stress in the hearts of NEK6 deficiency mice under diabetic cardiomyopathy pathology. Adenovirus was used to upregulate NEK6 in neonatal rat cardiomyocytes, and it was found that NEK6 ameliorated high glucose-induced inflammation and oxidative stress. Our findings revealed that NEK6 increased the phosphorylation of heat shock protein 72 (HSP72) and increased the protein level of PGC-1α and NRF2. Co-IP assay experiment confirmed that NEK6 interacted with HSP72. When HSP72 was silenced, the anti-inflammation and anti-oxidative stress effects of NEK6 were blurred. In summary, NEK6 may protect diabetic-induced cardiomyopathy by interacting with HSP72 and promoting the HSP72/PGC-1α/NRF2 signaling. KEY MESSAGES: NEK6 knockout deteriorated cardiac dysfunction, cardiac hypertrophy, fibrosis as well as inflammation response, and oxidative stress. NEK6 overexpression attenuated high glucose induced inflammation and oxidative stress. The underlying mechanisms of the protective role of NEK6 in the development of diabetic cardiomyopathy seem to involve the regulation of HSP72-NRF2- PGC-1α pathway. NEK6 may become a new therapeutic target for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Shuangyin Shao
- Department of Cardiovascular Surgery, Henan Provincial Chest Hospital, Zhengzhou University, Zhengzhou, 450000, China
| | - Lili Xiao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, China
| | - Meng Jia
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chuyang Zhang
- Department of Education, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guojun Zhao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, China
| | - Rui Yao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, China
| | - Xiaofang Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, China.
| | - Lu Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, China.
| |
Collapse
|
6
|
Zhu CS, Wang W, Qiang X, Chen W, Lan X, Li J, Wang H. Endogenous Regulation and Pharmacological Modulation of Sepsis-Induced HMGB1 Release and Action: An Updated Review. Cells 2021; 10:2220. [PMID: 34571869 PMCID: PMC8469563 DOI: 10.3390/cells10092220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/13/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis remains a common cause of death in intensive care units, accounting for approximately 20% of total deaths worldwide. Its pathogenesis is partly attributable to dysregulated inflammatory responses to bacterial endotoxins (such as lipopolysaccharide, LPS), which stimulate innate immune cells to sequentially release early cytokines (such as tumor necrosis factor (TNF) and interferons (IFNs)) and late mediators (such as high-mobility group box 1, HMGB1). Despite difficulties in translating mechanistic insights into effective therapies, an improved understanding of the complex mechanisms underlying the pathogenesis of sepsis is still urgently needed. Here, we review recent progress in elucidating the intricate mechanisms underlying the regulation of HMGB1 release and action, and propose a few potential therapeutic candidates for future clinical investigations.
Collapse
Affiliation(s)
- Cassie Shu Zhu
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; (C.S.Z.); (X.Q.); (W.C.); (X.L.); (J.L.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, USA
| | - Wei Wang
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA 24061, USA;
| | - Xiaoling Qiang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; (C.S.Z.); (X.Q.); (W.C.); (X.L.); (J.L.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, USA
| | - Weiqiang Chen
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; (C.S.Z.); (X.Q.); (W.C.); (X.L.); (J.L.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, USA
| | - Xiqian Lan
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; (C.S.Z.); (X.Q.); (W.C.); (X.L.); (J.L.)
| | - Jianhua Li
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; (C.S.Z.); (X.Q.); (W.C.); (X.L.); (J.L.)
| | - Haichao Wang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; (C.S.Z.); (X.Q.); (W.C.); (X.L.); (J.L.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, USA
| |
Collapse
|
7
|
Sahib S, Sharma A, Muresanu DF, Zhang Z, Li C, Tian ZR, Buzoianu AD, Lafuente JV, Castellani RJ, Nozari A, Patnaik R, Menon PK, Wiklund L, Sharma HS. Nanodelivery of traditional Chinese Gingko Biloba extract EGb-761 and bilobalide BN-52021 induces superior neuroprotective effects on pathophysiology of heat stroke. PROGRESS IN BRAIN RESEARCH 2021; 265:249-315. [PMID: 34560923 DOI: 10.1016/bs.pbr.2021.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Military personnel often exposed to high summer heat are vulnerable to heat stroke (HS) resulting in abnormal brain function and mental anomalies. There are reasons to believe that leakage of the blood-brain barrier (BBB) due to hyperthermia and development of brain edema could result in brain pathology. Thus, exploration of suitable therapeutic strategies is needed to induce neuroprotection in HS. Extracts of Gingko Biloba (EGb-761) is traditionally used in a variety of mental disorders in Chinese traditional medicine since ages. In this chapter, effects of TiO2 nanowired EGb-761 and BN-52021 delivery to treat brain pathologies in HS is discussed based on our own investigations. We observed that TiO2 nanowired delivery of EGb-761 or TiO2 BN-52021 is able to attenuate more that 80% reduction in the brain pathology in HS as compared to conventional drug delivery. The functional outcome after HS is also significantly improved by nanowired delivery of EGb-761 and BN-52021. These observations are the first to suggest that nanowired delivery of EGb-761 and BN-52021 has superior therapeutic effects in HS not reported earlier. The clinical significance in relation to the military medicine is discussed.
Collapse
Affiliation(s)
- Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Zhiqiang Zhang
- Department of Neurosurgery, Chinese Medicine Hospital of Guangdong Province, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Yuexiu, Guangzhou, China
| | - Cong Li
- Department of Neurosurgery, Chinese Medicine Hospital of Guangdong Province, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Yuexiu, Guangzhou, China
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Preeti K Menon
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
8
|
Min HJ, Kim KS, Choi GJ, Kang H, White FA. Concentrations of HMGB1 and Hsp70 of healthy subjects in upper and lower airway: Literature Review and Meta-analysis. Int J Med Sci 2021; 18:1760-1767. [PMID: 33746593 PMCID: PMC7976589 DOI: 10.7150/ijms.53500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 01/04/2021] [Indexed: 11/05/2022] Open
Abstract
Although high-mobility group box 1 and heat-shock protein 70 are implicated in airway diseases and suggested as relevant diagnostic biomarkers, their control concentrations in the airways have not yet been determined. This study aimed to evaluate concentration of healthy subjects for both these proteins in the upper and lower airways via meta-analysis. We searched MEDLINE, EMBASE, and Google Scholar for articles describing concentration of healthy subjects for these proteins. Data from healthy populations were combined using a random-effects model, and subgroup and sensitivity analyses were performed to determine between-study heterogeneity. We analyzed 22 studies involving 485 patients. Concentration of healthy subjects of high-mobility group box 1 and heat-shock protein 70 varied from "not detected" to 326.13 ng/mL and from 0.20 pg/mL to 9240.00 pg/mL, respectively, with the values showing significant heterogeneity. Subgroup analysis for high-mobility group box 1 revealed 13.63 ng/mL (95% CI 12.13-15.14), 100.31 ng/mL (95% CI -31.28-231.91), 9.54 ng/mL (95% CI 8.91-10.17), and 65.82 ng/mL (95% CI 55.51-76.14) for the lower airway, upper airway, pediatric populations, and adults, respectively, whereas that for heat-shock protein 70 revealed 20.58 pg/mL (95% CI 7.87-33.29) for the lower airway and 9240.00 ±11820 pg/mL for the upper airway. Although concentrations of healthy subjects of these proteins varied in the upper and lower airways, the levels of both these proteins were higher in the upper airway than in the lower airway, and these concentrations differed according to the age and sampling procedure. Our findings support the further evaluation of these proteins as biomarkers for airway-related diseases.
Collapse
Affiliation(s)
- Hyun Jin Min
- Department of Otorhinolaryngology-Head and Neck Surgery, Chung-Ang University College of Medicine, Seoul, Korea
| | - Kyung Soo Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Chung-Ang University College of Medicine, Seoul, Korea
| | - Geun Joo Choi
- Department of Anesthesiology and Pain Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Hyun Kang
- Department of Anesthesiology and Pain Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Fletcher A White
- Department of Anesthesia, Indiana University School of Medicine, IN, USA
| |
Collapse
|
9
|
Kim YH, Kwak MS, Lee B, Shin JM, Aum S, Park IH, Lee MG, Shin JS. Secretory autophagy machinery and vesicular trafficking are involved in HMGB1 secretion. Autophagy 2020; 17:2345-2362. [PMID: 33017561 PMCID: PMC8496717 DOI: 10.1080/15548627.2020.1826690] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Nuclear protein HMGB1 is secreted in response to various stimuli and functions as a danger-associated molecular pattern. Extracellular HMGB1 induces inflammation, cytokine production, and immune cell recruitment via activation of various receptors. As HMGB1 does not contain an endoplasmic reticulum-targeting signal peptide, HMGB1 is secreted via the endoplasmic reticulum-Golgi independently via an unconventional secretion pathway. However, the mechanism underlying HMGB1 secretion remains largely unknown. Here, we investigated the role of secretory autophagy machinery and vesicular trafficking in HMGB1 secretion. We observed that HSP90AA1 (heat shock protein 90 alpha family class A member 1), a stress-inducible protein, regulates the translocation of HMGB1 from the nucleus to the cytoplasm and its secretion through direct interaction. Additionally, geldanamycin, an HSP90AA1 inhibitor, reduced HMGB1 secretion. GORASP2/GRASP55 (golgi reassembly stacking protein 2), ARF1Q71L (ADP ribosylation factor 1), and SAR1AT39N (secretion associated Ras related GTPase 1A), which promoted unconventional protein secretion, increased HMGB1 secretion. HMGB1 secretion was inhibited by an early autophagy inhibitor and diminished in ATG5-deficient cells even when GORASP2 was overexpressed. In contrast, a late autophagy inhibitor increased HMGB1 secretion under the same conditions. The multivesicular body formation inhibitor GW4869 dramatically decreased HMGB1 secretion under HMGB1 secretion-inducing conditions. Thus, we demonstrated that secretory autophagy and multivesicular body formation mediate HMGB1 secretion.
Collapse
Affiliation(s)
- Young Hun Kim
- Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Man Sup Kwak
- Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Bin Lee
- Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Min Shin
- Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sowon Aum
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - In Ho Park
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Min Goo Lee
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea.,Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, Korea
| |
Collapse
|
10
|
Ryu SW, Stewart R, Pectol DC, Ender NA, Wimalarathne O, Lee JH, Zanini CP, Harvey A, Huibregtse JM, Mueller P, Paull TT. Proteome-wide identification of HSP70/HSC70 chaperone clients in human cells. PLoS Biol 2020; 18:e3000606. [PMID: 32687490 PMCID: PMC7392334 DOI: 10.1371/journal.pbio.3000606] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 07/30/2020] [Accepted: 06/29/2020] [Indexed: 12/25/2022] Open
Abstract
The 70 kDa heat shock protein (HSP70) family of chaperones are the front line of protection from stress-induced misfolding and aggregation of polypeptides in most organisms and are responsible for promoting the stability, folding, and degradation of clients to maintain cellular protein homeostasis. Here, we demonstrate quantitative identification of HSP70 and 71 kDa heat shock cognate (HSC70) clients using a ubiquitin-mediated proximity tagging strategy and show that, despite their high degree of similarity, these enzymes have largely nonoverlapping specificities. Both proteins show a preference for association with newly synthesized polypeptides, but each responds differently to changes in the stoichiometry of proteins in obligate multi-subunit complexes. In addition, expression of an amyotrophic lateral sclerosis (ALS)-associated superoxide dismutase 1 (SOD1) mutant protein induces changes in HSP70 and HSC70 client association and aggregation toward polypeptides with predicted disorder, indicating that there are global effects from a single misfolded protein that extend to many clients within chaperone networks. Together these findings show that the ubiquitin-activated interaction trap (UBAIT) fusion system can efficiently isolate the complex interactome of HSP chaperone family proteins under normal and stress conditions. Development of a ubiquitin-based system to comprehensively identify substrates of HSP70 enzymes in human cells reveals that constitutive HSC70 and stress-induced HSP70 have different binding preferences and respond dynamically to changes in misfolded protein levels.
Collapse
Affiliation(s)
- Seung W. Ryu
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Rose Stewart
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - D. Chase Pectol
- The Department of Chemistry, Texas A&M University, College Station, Texas, United States of America
| | - Nicolette A. Ender
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Oshadi Wimalarathne
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Ji-Hoon Lee
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Carlos P. Zanini
- Department of Statistics & Data Sciences, University of Texas at Austin, Austin, Texas, United States of America
| | - Antony Harvey
- Thermo Fisher Scientific, Austin, Texas, United States of America
| | - Jon M. Huibregtse
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Peter Mueller
- Department of Statistics & Data Sciences, University of Texas at Austin, Austin, Texas, United States of America
| | - Tanya T. Paull
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
11
|
Tang S, Du X, Yuan L, Xiao G, Wu M, Wang L, Wu S, Duan Z, Xiang Y, Qu X, Liu H, Zou Y, Qin X, Qin L, Liu C. Airway epithelial ITGB4 deficiency in early life mediates pulmonary spontaneous inflammation and enhanced allergic immune response. J Cell Mol Med 2020; 24:2761-2771. [PMID: 31970850 PMCID: PMC7077534 DOI: 10.1111/jcmm.15000] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 12/13/2019] [Accepted: 12/27/2019] [Indexed: 12/22/2022] Open
Abstract
Lung immune responses to respiratory pathogens and allergens are initiated in early life which will further influence the later onset of asthma. The airway epithelia form the first mechanical physical barrier to allergic stimuli and environmental pollutants, which is also the key regulator in the initiation and development of lung immune response. However, the epithelial regulation mechanisms of early-life lung immune responses are far from clear. Our previous study found that integrin β4 (ITGB4) is decreased in the airway epithelium of asthma patients with specific variant site. ITGB4 deficiency in adult mice aggravated the lung Th2 immune responses and enhanced airway hyper-responsiveness (AHR) with a house dust mite (HDM)-induced asthma model. However, the contribution of ITGB4 to the postnatal lung immune response is still obscure. Here, we further demonstrated that ITGB4 deficiency following birth mediates spontaneous lung inflammation with ILC2 activation and increased infiltration of eosinophils and lymphocytes. Moreover, ITGB4 deficiency regulated thymic stromal lymphopoietin (TSLP) production in airway epithelial cells through EGFR pathways. Neutralization of TSLP inhibited the spontaneous inflammation significantly in ITGB4-deficient mice. Furthermore, we also found that ITGB4 deficiency led to exaggerated lung allergic inflammation response to HDM stress. In all, these findings indicate that ITGB4 deficiency in early life causes spontaneous lung inflammation and induces exaggerated lung inflammation response to HDM aeroallergen.
Collapse
Affiliation(s)
- Sha Tang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, China
| | - Xizi Du
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Lin Yuan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Mengping Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Leyuan Wang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - ShuangYan Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Zhen Duan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Yizhou Zou
- Department of Immunology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Ling Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Chi Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China.,Research Center of China-Africa Infectious Diseases, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Zhang T, Gaffrey MJ, Thomas DG, Weber TJ, Hess BM, Weitz KK, Piehowski PD, Petyuk VA, Moore RJ, Qian WJ, Thrall BD. A proteome-wide assessment of the oxidative stress paradigm for metal and metal-oxide nanomaterials in human macrophages. NANOIMPACT 2020; 17:100194. [PMID: 32133426 PMCID: PMC7055704 DOI: 10.1016/j.impact.2019.100194] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Responsible implementation of engineered nanomaterials (ENMs) into commercial applications is an important societal issue, driving demand for new approaches for rapid and comprehensive evaluation of their bioactivity and safety. An essential part of any research focused on identifying potential hazards of ENMs is the appropriate selection of biological endpoints to evaluate. Herein, we use a tiered strategy employing both targeted biological assays and untargeted quantitative proteomics to elucidate the biological responses of human THP-1 derived macrophages across a library of metal/metal oxide ENMs, raised as priority ENMs for investigation by NIEHS's Nanomaterial Health Implications Research (NHIR) program. Our results show that quantitative cellular proteome profiles readily distinguish ENM types based on their cytotoxic potential according to induction of biological processes and pathways involved in the cellular antioxidant response, TCA cycle, oxidative stress, endoplasmic reticulum stress, and immune responses as major processes impacted. Interestingly, bioinformatics analysis of differentially expressed proteins also revealed new biological processes that were influenced by all ENMs independent of their cytotoxic potential. These included biological processes that were previously implicated as mechanisms cells employ as adaptive responses to low levels of oxidative stress, including cell adhesion, protein translation and protein targeting. Unsupervised clustering revealed the most striking proteome changes that differentiated ENM classes highlight a small subset of proteins involved in the oxidative stress response (HMOX1), protein chaperone functions (HS71B, DNJB1), and autophagy (SQSTM), providing a potential new panel of markers of ENM-induced cellular stress. To our knowledge, the results represent the most comprehensive profiling of the biological responses to a library of ENMs conducted using quantitative mass spectrometry-based proteomics. The results provide a basis to identify the patterns of a diverse set of cellular pathways and biological processes impacted by ENM exposure in an important immune cell type, laying the foundation for multivariate, pathway-level structure activity assessments of ENMs in the future.
Collapse
Affiliation(s)
- Tong Zhang
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA 99352
| | - Matthew J Gaffrey
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA 99352
| | - Dennis G Thomas
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA 99352
| | - Thomas J Weber
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA 99352
| | - Becky M Hess
- Signatures Sciences and Technology Division, Pacific Northwest National Laboratory, Richland, WA 99352
| | - Karl K Weitz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA 99352
| | - Paul D Piehowski
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA 99352
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA 99352
| | - Ronald J Moore
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA 99352
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA 99352
| | - Brian D Thrall
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland WA 99352
| |
Collapse
|
13
|
Kami K, Ohira T, Oishi Y, Nakajima T, Goto K, Ohira Y. Role of 72-kDa Heat Shock Protein in Heat-stimulated Regeneration of Injured Muscle in Rat. J Histochem Cytochem 2019; 67:791-799. [PMID: 31233366 DOI: 10.1369/0022155419859861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The regeneration of injured muscles is facilitated by intermittent heat stress. The 72-kDa heat shock protein (HSP72), the level of which is increased by heat stress, is likely involved in this effect, but the precise mechanism remains unclear. This study was conducted to investigate the localization and role(s) of HSP72 in the regenerating muscles in heat-stressed rats using immunohistochemistry. Heat stress was applied by immersion of the rat lower body into hot water (42C, 30 min, every other day) following injection of bupivacaine into the soleus muscles. After 1 week, we found that HSP72 was expressed at high levels not only in the surviving myofibers but also in the blood vessels of the regenerating muscles in heated rats. In addition, leukocytes, possibly granulocytes, expressing cluster of differentiation 43 within the blood capillaries surrounding the regenerating myofibers also highly expressed HSP72. In contrast, marked expression of HSP72 was not observed in the intact or regenerating muscles without heat stress. These results suggest that heat-stress-induced HSP72 within the myofibers, blood vessels, and circulating leukocytes may play important roles in enhancing regeneration of injured muscles by heat stress. Our findings would be useful to investigate cell-specific role(s) of HSP72 during skeletal muscle regeneration.
Collapse
Affiliation(s)
- Katsuya Kami
- Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takashi Ohira
- Division of Aerospace Medicine, Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan.,Research Center for Space and Medical Sciences, Doshisha University, Kyoto, Japan
| | - Yasuharu Oishi
- Faculty of Education, Kumamoto University, Kumamoto, Japan.,Research Center for Space and Medical Sciences, Doshisha University, Kyoto, Japan
| | - Takayuki Nakajima
- Department of Clinical Medicine, The Jikei University Hospital, Tokyo, Japan
| | - Katsumasa Goto
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Aichi, Japan.,Research Center for Space and Medical Sciences, Doshisha University, Kyoto, Japan
| | - Yoshinobu Ohira
- Graduate School of Medicine, Osaka University, Osaka, Japan.,Research Center for Space and Medical Sciences, Doshisha University, Kyoto, Japan
| |
Collapse
|
14
|
Ma KC, Schenck EJ, Pabon MA, Choi AMK. The Role of Danger Signals in the Pathogenesis and Perpetuation of Critical Illness. Am J Respir Crit Care Med 2019; 197:300-309. [PMID: 28977759 DOI: 10.1164/rccm.201612-2460pp] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Kevin C Ma
- 1 Division of Pulmonary and Critical Care Medicine and.,2 New York-Presbyterian Hospital, New York, New York
| | - Edward J Schenck
- 1 Division of Pulmonary and Critical Care Medicine and.,2 New York-Presbyterian Hospital, New York, New York
| | - Maria A Pabon
- 3 Division of General Internal Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York; and.,2 New York-Presbyterian Hospital, New York, New York
| | - Augustine M K Choi
- 1 Division of Pulmonary and Critical Care Medicine and.,2 New York-Presbyterian Hospital, New York, New York
| |
Collapse
|
15
|
den Dekker A, Davis FM, Kunkel SL, Gallagher KA. Targeting epigenetic mechanisms in diabetic wound healing. Transl Res 2019; 204:39-50. [PMID: 30392877 PMCID: PMC6331222 DOI: 10.1016/j.trsl.2018.10.001] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 10/02/2018] [Accepted: 10/02/2018] [Indexed: 02/07/2023]
Abstract
Impaired wound healing is a major secondary complication of type 2 diabetes that often results in limb loss and disability. Normal tissue repair progresses through discrete phases including hemostasis, inflammation, proliferation, and remodeling. In diabetes, normal progression through these phases is impaired resulting in a sustained inflammatory state and dysfunctional epithelialization in the wound. Due to their plasticity, macrophages play a critical role in the transition from the inflammation phase to the proliferation phase. Diabetes disrupts macrophage function by impairing monocyte recruitment to the wound, reducing phagocytosis, and prohibiting the transition of inflammatory macrophages to an anti-inflammatory state. Diabetes also impedes keratinocyte and fibroblast function during the later phases resulting in impaired epithelialization of the wound. Several recent studies suggest that altered epigenetic regulation of both immune and structural cells in wounds may influence cell phenotypes and healing, particularly in pathologic states, such as diabetes. Specifically, it has been shown that macrophage plasticity during wound repair is partly regulated epigenetically and that diabetes alters this epigenetic regulation and contributes to a sustained inflammatory state. Epigenetic regulation is also known to regulate keratinocyte and fibroblast function during wound repair. In this review, we provide an introduction to the epigenetic mechanisms that regulate tissue repair and highlight recent findings that demonstrate, how epigenetic events are altered during the course of diabetic wound healing.
Collapse
Affiliation(s)
- Aaron den Dekker
- Department of Surgery, University of Michigan, Ann Arbor, Michgan
| | - Frank M Davis
- Department of Surgery, University of Michigan, Ann Arbor, Michgan
| | - Steve L Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
16
|
Ulrich G, Salvadè A, Boersema P, Calì T, Foglieni C, Sola M, Picotti P, Papin S, Paganetti P. Phosphorylation of nuclear Tau is modulated by distinct cellular pathways. Sci Rep 2018; 8:17702. [PMID: 30531974 PMCID: PMC6286375 DOI: 10.1038/s41598-018-36374-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 11/16/2018] [Indexed: 11/28/2022] Open
Abstract
Post-translational protein modification controls the function of Tau as a scaffold protein linking a variety of molecular partners. This is most studied in the context of microtubules, where Tau regulates their stability as well as the distribution of cellular components to defined compartments. However, Tau is also located in the cell nucleus; and is found to protect DNA. Quantitative assessment of Tau modification in the nucleus when compared to the cytosol may elucidate how subcellular distribution and function of Tau is regulated. We undertook an unbiased approach by combing bimolecular fluorescent complementation and mass spectrometry in order to show that Tau phosphorylation at specific residues is increased in the nucleus of proliferating pluripotent neuronal C17.2 and neuroblastoma SY5Y cells. These findings were validated with the use of nuclear targeted Tau and subcellular fractionation, in particular for the phosphorylation at T181, T212 and S404. We also report that the DNA damaging drug Etoposide increases the translocation of Tau to the nucleus whilst reducing its phosphorylation. We propose that overt phosphorylation of Tau, a hallmark of neurodegenerative disorders defined as tauopathies, may negatively regulate the function of nuclear Tau in protecting against DNA damage.
Collapse
Affiliation(s)
- Giorgio Ulrich
- Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Ente Cantonale Ospedaliero, Torricella-Taverne, Switzerland
| | - Agnese Salvadè
- Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Ente Cantonale Ospedaliero, Torricella-Taverne, Switzerland
| | - Paul Boersema
- Institute of Molecular Systems Biology, Department of Biology, ETHZ, Zurich, Switzerland
| | - Tito Calì
- Department of Biomedical Sciences and Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Chiara Foglieni
- Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Ente Cantonale Ospedaliero, Torricella-Taverne, Switzerland
| | - Martina Sola
- Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Ente Cantonale Ospedaliero, Torricella-Taverne, Switzerland
| | - Paola Picotti
- Institute of Molecular Systems Biology, Department of Biology, ETHZ, Zurich, Switzerland
| | - Stéphanie Papin
- Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Ente Cantonale Ospedaliero, Torricella-Taverne, Switzerland
| | - Paolo Paganetti
- Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Ente Cantonale Ospedaliero, Torricella-Taverne, Switzerland.
| |
Collapse
|
17
|
Abstract
High-mobility group box 1 (HMGB1) is one of the most abundant proteins in eukaryotes and the best characterized damage-associated molecular pattern (DAMP). The biological activities of HMGB1 depend on its subcellular location, context and post-translational modifications. Inside the nucleus, HMGB1 is engaged in many DNA events such as DNA repair, transcription regulation and genome stability; in the cytoplasm, its main function is to regulate the autophagic flux while in the extracellular environment, it possesses more complicated functions and it is involved in a large variety of different processes such as inflammation, migration, invasion, proliferation, differentiation and tissue regeneration. Due to this pleiotropy, the role of HMGB1 has been vastly investigated in various pathological diseases and a large number of studies have explored its function in cardiovascular pathologies. However, in this contest, the precise mechanism of action of HMGB1 and its therapeutic potential are still very controversial since is debated whether HMGB1 is involved in tissue damage or plays a role in tissue repair and regeneration. The main focus of this review is to provide an overview of the effects of HMGB1 in different ischemic heart diseases and to discuss its functions in these pathological conditions.
Collapse
|
18
|
Mou K, Liu W, Miao Y, Cao F, Li P. HMGB1 deficiency reduces H 2 O 2 -induced oxidative damage in human melanocytes via the Nrf2 pathway. J Cell Mol Med 2018; 22:6148-6156. [PMID: 30338917 PMCID: PMC6237592 DOI: 10.1111/jcmm.13895] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/10/2018] [Accepted: 08/12/2018] [Indexed: 12/21/2022] Open
Abstract
Oxidative stress leads to melanocyte death and has been implicated in the pathogenesis of vitiligo. The nuclear factor, E2‐related factor 2 (Nrf2), is a critical transcription factor in protecting cells from oxidative damage. High‐mobility group box 1 (HMGB1) is a chromatin‐associated nuclear protein and an extracellular damage‐associated molecular pattern molecule. Extracellular HMGB1 released from activated immune cells, necrotic or injured cells, becomes a proinflammatory mediator through binding to cell‐surface receptors of responding cells. In this study, we investigated the role of HMGB1 from melanocytes in the response to oxidative stress and the mechanism involved. We showed that HMGB1 is expressed by primary normal human epidermal melanocytes (NHEMs). H2O2 treatment increased cytoplasmic translocation and extracellular release of HMGB1. HMGB1 knockdown by small interfering RNA (siRNA) led to decreased apoptosis of NHEMs. HMGB1 inhibition enhanced the expression of Nrf2 and its target genes. The expression of Nrf2 and its downstream antioxidant genes was downregulated after the supernatant of H2O2‐treated NHEMs was added to HMGB1‐deficient cells. HMGB1 knockdown by siRNA suppressed the expression of the autophagosome marker, LC3, and enhanced p62 expression. Coimmunoprecipitation with Keap1 showed a reduced Nrf2‐Keap1 interaction and an increased p62‐Keap1 interaction under oxidative stress. These data demonstrated that external stimuli (eg, oxidative stress) may trigger autocrine HMGB1 translocation and release by melanocytes, suppressing the expression of Nrf2 and downstream antioxidant genes to induce melanocyte apoptosis, and thereby participate in the pathological process of vitiligo.
Collapse
Affiliation(s)
- Kuanhou Mou
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wei Liu
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yi Miao
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fang Cao
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pan Li
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
19
|
Rao Y, Wan Q, Su H, Xiao X, Liao Z, Ji J, Yang C, Lin L, Su J. ROS-induced HSP70 promotes cytoplasmic translocation of high-mobility group box 1b and stimulates antiviral autophagy in grass carp kidney cells. J Biol Chem 2018; 293:17387-17401. [PMID: 30237170 DOI: 10.1074/jbc.ra118.003840] [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] [Received: 05/05/2018] [Revised: 09/11/2018] [Indexed: 12/20/2022] Open
Abstract
Autophagy plays many physiological and pathophysiological roles. However, the roles and the regulatory mechanisms of autophagy in response to viral infections are poorly defined in teleost fish, such as grass carp (Ctenopharyngodon idella), which is one of the most important aquaculture species in China. In this study, we found that both grass carp reovirus (GCRV) infection and hydrogen peroxide (H2O2) treatment induced the accumulation of reactive oxygen species (ROS) in C. idella kidney cells and stimulate autophagy. Suppressing ROS accumulation with N-acetyl-l-cysteine significantly inhibited GCRV-induced autophagy activation and enhanced GCRV replication. Although ROS-induced autophagy, in turn, restricted GCRV replication, further investigation revealed that the multifunctional cellular protein high-mobility group box 1b (HMGB1b) serves as a heat shock protein 70 (HSP70)-dependent, pro-autophagic protein in grass carp. Upon H2O2 treatment, cytoplasmic HSP70 translocated to the nucleus, where it interacted with HMGB1b and promoted cytoplasmic translocation of HMGB1b. Overexpression and siRNA-mediated knockdown assays indicated that HSP70 and HMGB1b synergistically enhance ROS-induced autophagic activation in the cytoplasm. Moreover, HSP70 reinforced an association of HMGB1b with the C. idella ortholog of Beclin 1 (a mammalian ortholog of the autophagy-associated yeast protein ATG6) by directly interacting with C. idella Beclin 1. In summary, this study highlights the antiviral function of ROS-induced autophagy in response to GCRV infection and reveals the positive role of HSP70 in HMGB1b-mediated autophagy initiation in teleost fish.
Collapse
Affiliation(s)
- Youliang Rao
- From the College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.,the Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Quanyuan Wan
- From the College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.,the College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China, and
| | - Hang Su
- From the College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Xun Xiao
- From the College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhiwei Liao
- From the College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Jianfei Ji
- From the College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Chunrong Yang
- the College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Li Lin
- the College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China, and
| | - Jianguo Su
- From the College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China, .,the Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|
20
|
Rosado MM, Simkó M, Mattsson MO, Pioli C. Immune-Modulating Perspectives for Low Frequency Electromagnetic Fields in Innate Immunity. Front Public Health 2018; 6:85. [PMID: 29632855 PMCID: PMC5879099 DOI: 10.3389/fpubh.2018.00085] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 03/05/2018] [Indexed: 12/21/2022] Open
Abstract
In recent years, the effects of electromagnetic fields (EMFs) on the immune system have received a considerable interest, not only to investigate possible negative health impact but also to explore the possibility to favorably modulate immune responses. To generate beneficial responses, the immune system should eradicate pathogens while “respecting” the organism and tolerating irrelevant antigens. According to the current view, damage-associated molecules released by infected or injured cells, or secreted by innate immune cells generate danger signals activating an immune response. These signals are also relevant to the subsequent activation of homeostatic mechanisms that control the immune response in pro- or anti-inflammatory reactions, a feature that allows modulation by therapeutic treatments. In the present review, we describe and discuss the effects of extremely low frequency (ELF)-EMF and pulsed EMF on cell signals and factors relevant to the activation of danger signals and innate immunity cells. By discussing the EMF modulating effects on cell functions, we envisage the use of EMF as a therapeutic agent to regulate immune responses associated with wound healing.
Collapse
Affiliation(s)
| | | | - Mats-Olof Mattsson
- AIT Austrian Institute of Technology, Center for Energy, Environmental Resources and Technologies, Tulln, Austria
| | - Claudio Pioli
- Laboratory of Biomedical Technologies, Division of Health Protection Technologies, ENEA, Rome, Italy
| |
Collapse
|
21
|
Zhou H, Li Y, Gui H, Zhao H, Wu M, Li G, Li Y, Bai Z, Yin Z, Redmond HP, Wang J, Wang JH, Zhao Z. Antagonism of Integrin CD11b Affords Protection against Endotoxin Shock and Polymicrobial Sepsis via Attenuation of HMGB1 Nucleocytoplasmic Translocation and Extracellular Release. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:1771-1780. [PMID: 29343555 DOI: 10.4049/jimmunol.1701285] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/15/2017] [Indexed: 09/13/2023]
Abstract
High mobility group box 1 (HMGB1), a chromatin-binding nuclear protein, plays a critical role in sepsis by acting as a key "late-phase" inflammatory mediator. Integrin CD11b is essential for inflammatory cell activation and migration, thus mediating inflammatory responses. However, it is unclear whether CD11b participates in the development of sepsis. In this study, we report that CD11b contributes to LPS-induced endotoxin shock and microbial sepsis, as antagonism of CD11b with the CD11b blocking Ab or CD11b inhibitor Gu-4 protects mice against LPS- and microbial sepsis-related lethality, which is associated with significantly diminished serum HMGB1 levels. Consistent with this, CD11b-deficient mice were more resistant to microbial sepsis with a much lower serum HMGB1 level compared with wild-type mice. Pharmacological blockage and genetic knockdown/knockout of CD11b in murine macrophages hampered LPS-stimulated HMGB1 nucleocytoplasmic translocation and extracellular release. Furthermore, silencing CD11b interrupted the interaction of HMGB1 with either a nuclear export factor chromosome region maintenance 1 or classical protein kinase C and inhibited classical protein kinase C-induced HMGB1 phosphorylation, the potential underlying mechanism(s) responsible for CD11b blockage-induced suppression of HMGB1 nucleocytoplasmic translocation and subsequent extracellular release. Thus, our results highlight that CD11b contributes to the development of sepsis, predominantly by facilitating nucleocytoplasmic translocation and active release of HMGB1.
Collapse
Affiliation(s)
- Huiting Zhou
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210023, China; and
| | - Yanhong Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Huan Gui
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - He Zhao
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Ming Wu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Gang Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Yiping Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Zhenjiang Bai
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210023, China; and
| | - H Paul Redmond
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork T12YN60, Ireland
| | - Jian Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China;
| | - Jiang Huai Wang
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork T12YN60, Ireland
| | - Zhihui Zhao
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210023, China; and
| |
Collapse
|
22
|
Regulation of Tumor Progression by Programmed Necrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3537471. [PMID: 29636841 PMCID: PMC5831895 DOI: 10.1155/2018/3537471] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/28/2017] [Indexed: 12/12/2022]
Abstract
Rapidly growing malignant tumors frequently encounter hypoxia and nutrient (e.g., glucose) deprivation, which occurs because of insufficient blood supply. This results in necrotic cell death in the core region of solid tumors. Necrotic cells release their cellular cytoplasmic contents into the extracellular space, such as high mobility group box 1 (HMGB1), which is a nonhistone nuclear protein, but acts as a proinflammatory and tumor-promoting cytokine when released by necrotic cells. These released molecules recruit immune and inflammatory cells, which exert tumor-promoting activity by inducing angiogenesis, proliferation, and invasion. Development of a necrotic core in cancer patients is also associated with poor prognosis. Conventionally, necrosis has been thought of as an unregulated process, unlike programmed cell death processes like apoptosis and autophagy. Recently, necrosis has been recognized as a programmed cell death, encompassing processes such as oncosis, necroptosis, and others. Metabolic stress-induced necrosis and its regulatory mechanisms have been poorly investigated until recently. Snail and Dlx-2, EMT-inducing transcription factors, are responsible for metabolic stress-induced necrosis in tumors. Snail and Dlx-2 contribute to tumor progression by promoting necrosis and inducing EMT and oncogenic metabolism. Oncogenic metabolism has been shown to play a role(s) in initiating necrosis. Here, we discuss the molecular mechanisms underlying metabolic stress-induced programmed necrosis that promote tumor progression and aggressiveness.
Collapse
|
23
|
van Eden W, Jansen MAA, Ludwig I, van Kooten P, van der Zee R, Broere F. The Enigma of Heat Shock Proteins in Immune Tolerance. Front Immunol 2017; 8:1599. [PMID: 29209330 PMCID: PMC5702443 DOI: 10.3389/fimmu.2017.01599] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/06/2017] [Indexed: 01/22/2023] Open
Abstract
The fundamental problem of autoimmune diseases is the failure of the immune system to downregulate its own potentially dangerous cells, which leads to destruction of tissue expressing the relevant autoantigens. Current immunosuppressive therapies offer relief but fail to restore the basic condition of self-tolerance. They do not induce long-term physiological regulation resulting in medication-free disease remissions. Heat shock proteins (HSPs) have shown to possess the capacity of inducing lasting protective immune responses in models of experimental autoimmune diseases. Especially mycobacterial HSP60 and HSP70 were shown to induce disease inhibitory IL-10-producing regulatory T cells in many different models. This in itself may seem enigmatic, since based on earlier studies, HSPs were also coined sometimes as pro-inflammatory damage-associated molecular patterns. First clinical trials with HSPs in rheumatoid arthritis and type I diabetes have also indicated their potential to restore tolerance in autoimmune diseases. Data obtained from the models have suggested three aspects of HSP as being critical for this tolerance promoting potential: 1. evolutionary conservation, 2. most frequent cytosolic/nuclear MHC class II natural ligand source, and 3. upregulation under (inflammatory) stress. The combination of these three aspects, which are each relatively unique for HSP, may provide an explanation for the enigmatic immune tolerance promoting potential of HSP.
Collapse
Affiliation(s)
- Willem van Eden
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine (FVM), Utrecht University, Utrecht, Netherlands
| | - Manon A A Jansen
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine (FVM), Utrecht University, Utrecht, Netherlands
| | - Irene Ludwig
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine (FVM), Utrecht University, Utrecht, Netherlands
| | - Peter van Kooten
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine (FVM), Utrecht University, Utrecht, Netherlands
| | - Ruurd van der Zee
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine (FVM), Utrecht University, Utrecht, Netherlands
| | - Femke Broere
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine (FVM), Utrecht University, Utrecht, Netherlands
| |
Collapse
|
24
|
Involvement of HSP70 and HO-1 in the protective effects of raloxifene on multiple organ dysfunction syndrome by endotoxemia in ovariectomized rats. Menopause 2017; 24:959-969. [DOI: 10.1097/gme.0000000000000864] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
25
|
Ren L, Tao J, Chen H, Bian Y, Yang X, Chen G, Zhang X, Liang G, Wu W, Song Z, Wang Y. Myeloid differentiation protein 2-dependent mechanisms in retinal ischemia-reperfusion injury. Toxicol Appl Pharmacol 2017; 317:1-11. [DOI: 10.1016/j.taap.2017.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/29/2016] [Accepted: 01/03/2017] [Indexed: 12/22/2022]
|
26
|
Abstract
Danger-associated molecular patterns (DAMPs) that are released by injured, threatened, or dead cells, or that originate from the extracellular matrix, influence the immune system. This is of great relevance in critically ill patients, in whom trauma or surgery-related cell damage, hypoxia, ischemia, and infections can result in extensive release of DAMPs. As many patients at the intensive care unit suffer from immune system-related complications, DAMPs could serve as markers for the prognosis of these patients and represent possible therapeutic targets. In the present review, we provide an overview of several well known DAMPs (high-mobility group box 1, heat-shock proteins, s100 proteins, nucleic acids, and hyaluronan) and their effects on the immune system. Furthermore, we discuss the role of DAMPs as markers or therapeutic targets in several conditions frequently encountered in critically ill patients, such as sepsis, trauma, ventilator-induced lung injury, and cardiac arrest.
Collapse
|
27
|
Minutti CM, Knipper JA, Allen JE, Zaiss DMW. Tissue-specific contribution of macrophages to wound healing. Semin Cell Dev Biol 2016; 61:3-11. [PMID: 27521521 DOI: 10.1016/j.semcdb.2016.08.006] [Citation(s) in RCA: 315] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/08/2016] [Accepted: 08/09/2016] [Indexed: 12/21/2022]
Abstract
Macrophages are present in all tissues, either as resident cells or monocyte-derived cells that infiltrate into tissues. The tissue site largely determines the phenotype of tissue-resident cells, which help to maintain tissue homeostasis and act as sentinels of injury. Both tissue resident and recruited macrophages make a substantial contribution to wound healing following injury. In this review, we evaluate how macrophages in two fundamentally distinct tissues, i.e. the lung and the skin, differentially contribute to the process of wound healing. We highlight the commonalities of macrophage functions during repair and contrast them with distinct, tissue-specific functions that macrophages fulfill during the different stages of wound healing.
Collapse
Affiliation(s)
- Carlos M Minutti
- Centre for Immunity, Infection and Evolution, and the Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, United Kingdom
| | - Johanna A Knipper
- Centre for Immunity, Infection and Evolution, and the Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, United Kingdom
| | - Judith E Allen
- School of Biological Sciences, Faculty of Biology, Medicine & Health & Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, United Kingdom.
| | - Dietmar M W Zaiss
- Centre for Immunity, Infection and Evolution, and the Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, United Kingdom.
| |
Collapse
|
28
|
Tang Y, Zhao X, Antoine D, Xiao X, Wang H, Andersson U, Billiar TR, Tracey KJ, Lu B. Regulation of Posttranslational Modifications of HMGB1 During Immune Responses. Antioxid Redox Signal 2016; 24:620-34. [PMID: 26715031 PMCID: PMC5349223 DOI: 10.1089/ars.2015.6409] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
SIGNIFICANCE High-mobility group protein 1 (HMGB1) is an evolutionarily conserved and multifunctional protein. The biological function of HMGB1 depends on its cellular locations, binding partners, and redox states. Extracellular HMGB1 is a mediator of inflammation during infection or tissue injury. Immune cells actively release HMGB1 in response to infection, which in turn orchestrates both innate and adaptive immune responses. RECENT ADVANCES Hyperacetylation of HMGB1 within its nuclear localization sequences mobilizes HMGB1 from the nucleus to the cytoplasm and subsequently promotes HMGB1 release. The redox states of the cysteines in positions 23, 45, and 106 determine the biological activity of the extracellular HMGB1. CRITICAL ISSUES The full picture and the detailed molecular mechanisms of how cells regulate the posttranslational modifications and the redox status of HMGB1 during immune responses or under stress not only unravel the molecular mechanisms by which cells regulate the release and the biological function of HMGB1 but may also provide novel therapeutic targets to treat inflammatory diseases. FUTURE DIRECTIONS It is important to identify the signaling pathways that regulate the posttranslational modifications and the redox status of HMGB1 and find their roles in host immune responses and pathogenesis of diseases.
Collapse
Affiliation(s)
- Yiting Tang
- 1 Department of Hematology, The 3rd Xiangya Hospital, Central South University , Changsha, China .,2 State Key Laboratory of Medical Genetics, School of Biological Science and Technology, Central South University , Changsha, China .,3 Department of Physiology, Xiangya School of Medicine, Central South University , Changsha, China
| | - Xin Zhao
- 1 Department of Hematology, The 3rd Xiangya Hospital, Central South University , Changsha, China .,2 State Key Laboratory of Medical Genetics, School of Biological Science and Technology, Central South University , Changsha, China
| | - Daniel Antoine
- 4 Department of Molecular and Clinical Pharmacology, Medical Research Council Centre for Drug Safety Science, University of Liverpool , Liverpool, United Kingdom
| | - Xianzhong Xiao
- 5 Hunan Province Key Laboratory of Sepsis and Translational Medicine, Xiangya School of Medicine, Central South University , Changsha, China .,6 Department of Pathophysiology, Xiangya School of Medicine, Central South University , Changsha, China
| | - Haichao Wang
- 7 Department of Emergency Medicine, North Shore University Hospital , Manhasset, New York
| | - Ulf Andersson
- 8 Department of Women's and Children's Health, Karolinska Institutet , Stockholm, Sweden
| | - Timothy R Billiar
- 9 Department of Surgery, University of Pittsburgh Medical Center , Pittsburgh, Pennsylvania
| | - Kevin J Tracey
- 10 Center of Biomedical Science, Feinstein Institute for Medical Research , Manhasset, New York
| | - Ben Lu
- 1 Department of Hematology, The 3rd Xiangya Hospital, Central South University , Changsha, China .,2 State Key Laboratory of Medical Genetics, School of Biological Science and Technology, Central South University , Changsha, China .,5 Hunan Province Key Laboratory of Sepsis and Translational Medicine, Xiangya School of Medicine, Central South University , Changsha, China .,10 Center of Biomedical Science, Feinstein Institute for Medical Research , Manhasset, New York
| |
Collapse
|
29
|
Liu XX, Wang C, Huang SF, Chen Q, Hu YF, Zhou L, Gu Y. Regnase-1 in microglia negatively regulates high mobility group box 1-mediated inflammation and neuronal injury. Sci Rep 2016; 6:24073. [PMID: 27044405 PMCID: PMC4820713 DOI: 10.1038/srep24073] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 03/18/2016] [Indexed: 01/04/2023] Open
Abstract
Extracellular high mobility group box 1 (HMGB1) has been demonstrated to function as a proinflammatory cytokine and induces neuronal injury in response to various pathological stimuli in central nervous system (CNS). However, the regulatory factor involved in HMGB1-mediated inflammatory signaling is largely unclear. Regulatory RNase 1 (Regnase-1) is a potent anti-inflammation enzyme that can degrade a set of mRNAs encoding proinflammatory cytokines. The present study aims to determine the role of Regnase-1 in the regulation of HMGB1-mediated inflammatory injury in CNS. Cultured microglia and rat brain were treated with recombinant HMGB1 to examine the induction of Regnase-1 expression. Moreover, the role of Regnase-1 in modulating the expression of inflammatory cytokines and neuronal injury was then investigated in microglia by specific siRNA knockdown upon HMGB1 treatment. Results showed that HMGB1 could significantly induce the de novo synthesis of Regnase-1 in cultured microglia. Consistently, Regnase-1 was elevated and found to be co-localized with microglia marker in the brain of rat treated with HMGB1. Silencing Regnase-1 in microglia enhanced HMGB1-induced expression of proinflammatory cytokines and exacerbated neuronal toxicity. Collectively, these results suggest that Regnase-1 can be induced by HMGB1 in microglia and negatively regulates HMGB1-mediated neuroinflammation and neuronal toxicity.
Collapse
Affiliation(s)
- Xiao-Xi Liu
- Department of Neurology, Nanfang Hospital, Southern Medical University. Guangzhou, Guangdong 510515, P. R. China
| | - Chen Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University. Guangzhou, Guangdong 510515, P. R. China.,Department of Neurology, the Second Affiliated Hospital &Yuying Children's Hospital, Wenzhou Medical University. Wenzhou, Zhejiang 325000, P. R. China
| | - Shao-Fei Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University. Guangzhou, Guangdong 510515, P. R. China
| | - Qiong Chen
- Department of Neurology, Nanfang Hospital, Southern Medical University. Guangzhou, Guangdong 510515, P. R. China
| | - Ya-Fang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University. Guangzhou, Guangdong 510515, P. R. China
| | - Liang Zhou
- Department of Neurology, Nanfang Hospital, Southern Medical University. Guangzhou, Guangdong 510515, P. R. China
| | - Yong Gu
- Department of Neurology, Nanfang Hospital, Southern Medical University. Guangzhou, Guangdong 510515, P. R. China
| |
Collapse
|
30
|
Wang L, He L, Bao G, He X, Fan S, Wang H. Ionizing Radiation Induces HMGB1 Cytoplasmic Translocation and Extracellular Release. GUO JI FANG SHE YI XUE HE YI XUE ZA ZHI = INTERNATIONAL JOURNAL OF RADIATION MEDICINE AND NUCLEAR MEDICINE 2016; 40:91-99. [PMID: 27331198 PMCID: PMC4911189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
OBJECTIVE A nucleosomal protein, HMGB1, can be secreted by activated immune cells or passively released by dying cells, thereby amplifying rigorous inflammatory responses. In this study we aimed to test the possibility that ionizing radiation similarly induces cytoplasmic HMGB1 translocation and extracellular release. METHOD Human skin fibroblast (GM0639) and bronchial epithelial (16HBE) cells and animals (rats) were exposed to X-ray radiation, and HMGB1 translocation and release were assessed by immunocytochemistry and immunoassay, respectively. RESULTS At a wide dose range (4.0 - 12.0 Gy), X-ray radiation induced a dramatic cytoplasmic HMGB1 translocation, and triggered a time- and dose-dependent HMGB1 release both in vitro and in vivo. The radiation-mediated HMGB1 release was associated with noticeable chromosomal DNA damage and loss of cell viability. CONCLUSION radiation induces HMGB1 cytoplasmic translocation and extracellular release through active secretion and passive leakage processes.
Collapse
Affiliation(s)
- Lili Wang
- School of Radiation and Public Health, Soochow University Medical College, Suzhou, Jiangsu 215123, China
| | - Li He
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Guoqiang Bao
- Department of General Surgery, Tangdu Hospital, The 4 Military Medical University, Xi’an, Shaanxi, 710032, China
- Laboratory of Emergency Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Xin He
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Saijun Fan
- School of Radiation and Public Health, Soochow University Medical College, Suzhou, Jiangsu 215123, China
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Haichao Wang
- School of Radiation and Public Health, Soochow University Medical College, Suzhou, Jiangsu 215123, China
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
- Laboratory of Emergency Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| |
Collapse
|
31
|
Bukar Maina M, Al-Hilaly YK, Serpell LC. Nuclear Tau and Its Potential Role in Alzheimer's Disease. Biomolecules 2016; 6:9. [PMID: 26751496 PMCID: PMC4808803 DOI: 10.3390/biom6010009] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 12/16/2015] [Accepted: 12/23/2015] [Indexed: 12/14/2022] Open
Abstract
Tau protein, found in both neuronal and non-neuronal cells, forms aggregates in neurons that constitutes one of the hallmarks of Alzheimer’s disease (AD). For nearly four decades, research efforts have focused more on tau’s role in physiology and pathology in the context of the microtubules, even though, for over three decades, tau has been localised in the nucleus and the nucleolus. Its nuclear and nucleolar localisation had stimulated many questions regarding its role in these compartments. Data from cell culture, mouse brain, and the human brain suggests that nuclear tau could be essential for genome defense against cellular distress. However, its nature of translocation to the nucleus, its nuclear conformation and interaction with the DNA and other nuclear proteins highly suggest it could play multiple roles in the nucleus. To find efficient tau-based therapies, there is a need to understand more about the functional relevance of the varied cellular distribution of tau, identify whether specific tau transcripts or isoforms could predict tau’s localisation and function and how they are altered in diseases like AD. Here, we explore the cellular distribution of tau, its nuclear localisation and function and its possible involvement in neurodegeneration.
Collapse
Affiliation(s)
- Mahmoud Bukar Maina
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, East Sussex, UK.
- Department of Human Anatomy, College of Medical Science, Gombe State University, Gombe 760, Nigeria.
| | - Youssra K Al-Hilaly
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, East Sussex, UK.
- Chemistry Department, College of Sciences, Al-Mustansiriyah University, Baghdad, Iraq.
| | - Louise C Serpell
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, East Sussex, UK.
| |
Collapse
|
32
|
Huang J, Xie Y, Sun X, Zeh HJ, Kang R, Lotze MT, Tang D. DAMPs, ageing, and cancer: The 'DAMP Hypothesis'. Ageing Res Rev 2015; 24:3-16. [PMID: 25446804 DOI: 10.1016/j.arr.2014.10.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 09/24/2014] [Accepted: 10/22/2014] [Indexed: 12/25/2022]
Abstract
Ageing is a complex and multifactorial process characterized by the accumulation of many forms of damage at the molecular, cellular, and tissue level with advancing age. Ageing increases the risk of the onset of chronic inflammation-associated diseases such as cancer, diabetes, stroke, and neurodegenerative disease. In particular, ageing and cancer share some common origins and hallmarks such as genomic instability, epigenetic alteration, aberrant telomeres, inflammation and immune injury, reprogrammed metabolism, and degradation system impairment (including within the ubiquitin-proteasome system and the autophagic machinery). Recent advances indicate that damage-associated molecular pattern molecules (DAMPs) such as high mobility group box 1, histones, S100, and heat shock proteins play location-dependent roles inside and outside the cell. These provide interaction platforms at molecular levels linked to common hallmarks of ageing and cancer. They can act as inducers, sensors, and mediators of stress through individual plasma membrane receptors, intracellular recognition receptors (e.g., advanced glycosylation end product-specific receptors, AIM2-like receptors, RIG-I-like receptors, and NOD1-like receptors, and toll-like receptors), or following endocytic uptake. Thus, the DAMP Hypothesis is novel and complements other theories that explain the features of ageing. DAMPs represent ideal biomarkers of ageing and provide an attractive target for interventions in ageing and age-associated diseases.
Collapse
|
33
|
Marino LV, Pathan N, Meyer RW, Wright VJ, Habibi P. An in vitro model to consider the effect of 2 mM glutamine and KNK437 on endotoxin-stimulated release of heat shock protein 70 and inflammatory mediators. Nutrition 2015; 32:375-83. [PMID: 26706024 DOI: 10.1016/j.nut.2015.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/13/2015] [Accepted: 09/13/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Glutamine has been shown to promote the release of heat shock protein 70 (HSP70) both within experimental in vitro models of sepsis and in adults with septic shock. This study aimed to investigate the effects of 2 mM glutamine and an inhibitor of HSP70 (KNK437) on the release of HSP70 and inflammatory mediators in healthy adult volunteers. METHODS An in vitro whole blood endotoxin stimulation assay was used. RESULTS The addition of 2 mM glutamine significantly increased HSP70 levels over time (P < 0.05). HSP70 release had a positive correlation at 4 h with IL-1 β (r = 0.51, P = 0.03) and an inverse correlation with TNF-α (r = -0.56, P = 0.02) and IL-8 levels (r = -0.52, P = 0.03), and there were no significant correlations between HSP70 and IL6 or IL-10 or glutamine. Glutamine supplementation significantly (P < 0.05) attenuated the release of IL-10 at 4 h and IL-8 at 24 h, compared with conditions without glutamine. In endotoxin-stimulated blood there were no significant differences in the release of IL-6, TNF-α, and IL-1 β with glutamine supplementation at 4 and 24 h. However, glutamine supplementation (2 mM) appeared to attenuate the release of inflammatory mediators (IL-1 β, IL-6, TNF-α), although this effect was not statistically significant. The addition of KNK437, a HSP70 inhibitor, significantly diminished HSP70 release, which resulted in lower levels of inflammatory mediators (P < 0.05). CONCLUSION Glutamine supplementation promotes HSP70 release in an experimental model of sepsis. After the addition of KNK437, the effects of glutamine on HSP70 and inflammatory mediator release appear to be lost, suggesting that HSP70 in part orchestrates the inflammatory mediator response to sepsis. The clinical implications require further investigation.
Collapse
Affiliation(s)
- Luise V Marino
- Department of Paediatrics, Imperial College London, London, United Kingdom.
| | - Nazima Pathan
- Department of Paediatrics, School of Clinical Medicine, Cambridge University, Cambridge, United Kingdom
| | - Rosan W Meyer
- Department of Gastroenterology, Great Ormond Street Hospital for Sick Children, London, United Kingdom
| | - Victoria J Wright
- Department of Paediatrics, Imperial College London, London, United Kingdom
| | - Parviz Habibi
- Department of Paediatrics, Imperial College London, London, United Kingdom
| |
Collapse
|
34
|
Short-term heat pre-treatment modulates the release of HMGB1 and pro-inflammatory cytokines in hPDL cells following mechanical loading and affects monocyte behavior. Clin Oral Investig 2015; 20:923-31. [PMID: 26358476 DOI: 10.1007/s00784-015-1580-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 08/26/2015] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Heat shock proteins (HSP) act as cell-protective molecules that are upregulated upon thermal insult, hypoxia, and ischemia. Such ischemic conditions can be found during tissue remodeling associated with orthodontic tooth movement or trauma when compression forces lead to cell necrosis and subsequent clearance of cellular debris by immune competent cells. Host immune overreaction can result in undesired side effects such as tooth root resorption. Here, we analyzed whether heat pre-treatment would affect the initially catabolic host immune response induced by mechanical loading of human periodontal ligament (hPDL) cells, which represent major constituents of the tooth supporting apparatus involved in the regulation of periodontal remodeling. MATERIALS AND METHODS Fifth passage hPDL cells were exposed to an elevated temperature of 43° for 1 h prior to mechanical loading. Cell morphology, high mobility group box protein 1 (HMGB1), interleukin (IL)-6, and IL-8 expression were analyzed microscopically and by ELISA. The physiological relevance for monocyte behavior was tested in monocyte adhesion and osteoclast differentiation assays. RESULTS Short-term heat pre-treatment did not show any visible effect on hPDL cell morphology, but resulted in a significant downregulation of pro-inflammatory cytokines when being additionally loaded mechanically. Supernatants of heat-exposed hPDL cell cultures demonstrated a reduced impact on monocyte adhesion and osteoclastic differentiation. CONCLUSIONS Heat pre-treatment of hPDL cells induces cell-protective mechanisms towards mechanical stress and favors the reduction of cell stress associated effects on monocyte/macrophage physiology. CLINICAL RELEVANCE These data present the induction of heat shock proteins as a promising treatment option to limit undesired side effects of periodontal remodeling.
Collapse
|
35
|
Kong Q, Xu LH, Xu W, Fang JP, Xu HG. HMGB1 translocation is involved in the transformation of autophagy complexes and promotes chemoresistance in leukaemia. Int J Oncol 2015; 47:161-70. [PMID: 25954971 DOI: 10.3892/ijo.2015.2985] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 04/17/2015] [Indexed: 11/06/2022] Open
Abstract
Acute lymphoblastic leukaemia (ALL) is a common paediatric cancer and is among the most curable cancers. However, the acquisition of drug resistance is a significant obstacle to the achievement of favourable outcomes, and autophagy is regarded as a mechanism that underlies chemoresistance. In this study, RT-qPCR was used to measure the expression of HMGB1 and Beclin1 in bone marrow mononuclear cells. A CCK-8 test was conducted to assess cell viability. Western blot, immunofluorescence and transmission electron microscopic analyses were performed to evaluate the autophagy levels. Immunoprecipitation analysis was performed to detect protein-protein interactions in the autophagy complexes. We found that HMGB1 expression correlated with the clinical status of ALL. In vitro, anticancer agent-induced cytotoxic effects were associated with autophagy-related drug resistance, and these effects were ameliorated by FIP200 depletion or the application of autophagy inhibitors. Moreover, the Ulk1‑Atg13-FIP200 complex, which promotes HMGB1 trafficking, acted upstream of the HMGB1-Beclin1 and PI3KC3-Beclin1 complexes and played a critical role in autophagy. Targeting the transformation of autophagic complexes or HMGB1 translocation may suppress autophagy and consequently overcome chemoresistance in leukaemia.
Collapse
Affiliation(s)
- Qian Kong
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Lu-Hong Xu
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Wei Xu
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Jian-Pei Fang
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Hong-Gui Xu
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
36
|
Abstract
High mobility group box 1 (HMGB1) is a widely-expressed and highly-abundant protein that acts as an extracellular signal upon active secretion by immune cells or passive release by dead, dying, and injured cells. Both intracellular and extracellular HMGB1 play pivotal roles in regulation of the cellular response to stress. Targeting the translocation, release, and activity of HMGB1 can limit inflammation and reduce tissue damage during infection and sterile inflammation. Although the mechanisms contributing to HMGB1 biology are still under investigation, it appears that oxidative stress is a central regulator of HMGB1's translocation, release, and activity in inflammation and cell death (e.g., necrosis, apoptosis, autophagic cell death, pyroptosis, and NETosis). Thus, targeting HMGB1 with antioxidant compounds may be an attractive therapeutic strategy for inflammation-associated diseases such as sepsis, ischemia and reperfusion injury, arthritis, diabetes, and cancer.
Collapse
Affiliation(s)
- Yan Yu
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh Pittsburgh, PA, USA
| | - Daolin Tang
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh Pittsburgh, PA, USA
| | - Rui Kang
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh Pittsburgh, PA, USA
| |
Collapse
|
37
|
Lu X, Bi YW, Chen KB, Wang HY. Protective effect of olmesartan against cardiac ischemia/reperfusion injury in spontaneously hypertensive rats. Exp Ther Med 2015; 9:2081-2087. [PMID: 26136939 DOI: 10.3892/etm.2015.2373] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 03/05/2015] [Indexed: 01/31/2023] Open
Abstract
Olmesartan, as a new angiotensin II receptor blocker, has shown beneficial effects on cardiovascular diseases. Nevertheless, the effect of olmesartan on ischemia/reperfusion (I/R) injury in the hypertensive heart has not been investigated. Therefore, the present study aimed to investigate the effect of olmesartan on I/R injury in spontaneously hypertensive rats (SHRs). Experimental groups were designed with a 2×2 factorial design for olmesartan and I/R effects. In the I/R group, the left anterior descending coronary artery (LAD) was ligated for 40 min followed by a 180-min reperfusion. In the sham group, SHRs underwent the same surgical procedure as the I/R group, with the exception that the suture passed under the LAD without being tightened. In the Olm-I/R group, the SHRs received olmesartan (5 mg/kg) for 4 weeks prior to surgery and other procedures were the same as for the I/R group. In the Olm-sham group, the SHRs received olmesartan (5 mg/kg) for 4 weeks prior to surgery and other procedures were the same as for the sham group. Infarct size was measured for the I/R and Olm-I/R groups. Blood pressure (BP), serum creatine kinase (CK), left ventricular mass index (LVMI), high mobility group box 1 (HMGB1) protein expression levels and hypoxia-inducible factor-1α (HIF-1α) mRNA expression levels were measured for all four groups. Olmesartan significantly reduced BP and LVMI in the olmesartan-treated SHRs compared with those in the SHRs that were not treated with olmesartan. HMGB1 and HIF-1α expression levels were significantly decreased in the Olm-sham and Olm-I/R groups compared with those in the sham and I/R groups, respectively. The proportional increase in HIF-1α expression due to I/R was greater in the olmesartan-treated rats than in the untreated rats. Serum CK levels were significantly reduced in the Olm-I/R group compared with those in the I/R group. In conclusion, olmesartan ameliorates left ventricular hypotrophy and protects the heart against I/R injury in addition to lowing BP in SHRs. The protective effect of olmesartan may be partly due to its antioxidative and anti-inflammatory properties.
Collapse
Affiliation(s)
- Xin Lu
- Department of Cardiovascular Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China ; Department of Cardiovascular Surgery, Taian City Central Hospital, Taian, Shandong 271000, P.R. China
| | - Yan-Wen Bi
- Department of Cardiovascular Surgery, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Ke-Biao Chen
- Department of Cardiovascular Surgery, Taian City Central Hospital, Taian, Shandong 271000, P.R. China
| | - Hong-Yue Wang
- Department of Cardiovascular Surgery, Taian City Central Hospital, Taian, Shandong 271000, P.R. China
| |
Collapse
|
38
|
Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, Sun X, Wang H, Wang Q, Tsung A, Billiar TR, Zeh HJ, Lotze MT, Tang D. HMGB1 in health and disease. Mol Aspects Med 2014; 40:1-116. [PMID: 25010388 PMCID: PMC4254084 DOI: 10.1016/j.mam.2014.05.001] [Citation(s) in RCA: 731] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/05/2014] [Indexed: 12/22/2022]
Abstract
Complex genetic and physiological variations as well as environmental factors that drive emergence of chromosomal instability, development of unscheduled cell death, skewed differentiation, and altered metabolism are central to the pathogenesis of human diseases and disorders. Understanding the molecular bases for these processes is important for the development of new diagnostic biomarkers, and for identifying new therapeutic targets. In 1973, a group of non-histone nuclear proteins with high electrophoretic mobility was discovered and termed high-mobility group (HMG) proteins. The HMG proteins include three superfamilies termed HMGB, HMGN, and HMGA. High-mobility group box 1 (HMGB1), the most abundant and well-studied HMG protein, senses and coordinates the cellular stress response and plays a critical role not only inside of the cell as a DNA chaperone, chromosome guardian, autophagy sustainer, and protector from apoptotic cell death, but also outside the cell as the prototypic damage associated molecular pattern molecule (DAMP). This DAMP, in conjunction with other factors, thus has cytokine, chemokine, and growth factor activity, orchestrating the inflammatory and immune response. All of these characteristics make HMGB1 a critical molecular target in multiple human diseases including infectious diseases, ischemia, immune disorders, neurodegenerative diseases, metabolic disorders, and cancer. Indeed, a number of emergent strategies have been used to inhibit HMGB1 expression, release, and activity in vitro and in vivo. These include antibodies, peptide inhibitors, RNAi, anti-coagulants, endogenous hormones, various chemical compounds, HMGB1-receptor and signaling pathway inhibition, artificial DNAs, physical strategies including vagus nerve stimulation and other surgical approaches. Future work further investigating the details of HMGB1 localization, structure, post-translational modification, and identification of additional partners will undoubtedly uncover additional secrets regarding HMGB1's multiple functions.
Collapse
Affiliation(s)
- Rui Kang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| | - Ruochan Chen
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Qiuhong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Wen Hou
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Sha Wu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Lizhi Cao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yan Yu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xue-Gong Fan
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhengwen Yan
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA; Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Xiaofang Sun
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Experimental Department of Institute of Gynecology and Obstetrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510510, China
| | - Haichao Wang
- Laboratory of Emergency Medicine, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Qingde Wang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Herbert J Zeh
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Daolin Tang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| |
Collapse
|
39
|
Innovative Diagnostik für die pädiatrische Infektiologie. Monatsschr Kinderheilkd 2014. [DOI: 10.1007/s00112-014-3100-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
40
|
In vitro palmitate treatment of myotubes from postmenopausal women leads to ceramide accumulation, inflammation and affected insulin signaling. PLoS One 2014; 9:e101555. [PMID: 25000528 PMCID: PMC4084812 DOI: 10.1371/journal.pone.0101555] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 06/07/2014] [Indexed: 11/19/2022] Open
Abstract
Menopause is associated with an increased incidence of insulin resistance and metabolic diseases. In a chronic palmitate treatment model, we investigated the role of skeletal muscle fatty acid exposure in relation to the metabolic deterioration observed with menopause. Human skeletal muscle satellite cells were isolated from premenopausal (n = 6) and postmenopausal (n = 5) women. In an in vitro model, the myotubes were treated with palmitate (300 µM) for one-, two- or three days during differentiation. Effects on lipid accumulation, inflammation and insulin signaling were studied. Palmitate treatment led to a 108% (CI 95%: 50%; 267%) increase in intramyocellular ceramide in the myotubes from the postmenopausal women (post-myotubes) compared with a 26% (CI 95%: -57%; 96%) increase in myotubes from the premenopausal women (pre-myotubes), (p<0.05). Furthermore, post-myotubes had a 22% (CI 95%: 4%; 34%) increase in pJNK (p = 0.04) and a 114% (CI 95%: 50%; 177%) increase in Hsp70 protein expression (p = 0.03) after three days of palmitate treatment, compared with pre-myotubes, in which no increase in either pJNK (-12% (CI 95: -26%; 2%)) or Hsp70 (7% (CI 95: -78%; 91%)) was detected. Furthermore, post-myotubes showed a blunted insulin stimulated phosphorylation of AS160 in response to chronic palmitate treatment compared with pre-myotubes (p = 0.02). The increased intramyocellular ceramide content in the post-myotubes was associated with a significantly higher mRNA expression of Serine Palmitoyltransferase1 (SPT1) after one day of palmitate treatment (p = 0.03) in post-myotubes compared with pre-myotubes. Our findings indicate that post-myotubes are more prone to develop lipid accumulation and defective insulin signaling following chronic saturated fatty acid exposure as compared to pre-myotubes.
Collapse
|
41
|
Wang K, Lei J, Zou J, Xiao H, Chen A, Liu X, Liu Y, Jiang L, Xiao Z, Xiao X. Mipu1, a novel direct target gene, is involved in hypoxia inducible factor 1-mediated cytoprotection. PLoS One 2013; 8:e82827. [PMID: 24349374 PMCID: PMC3859620 DOI: 10.1371/journal.pone.0082827] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 11/06/2013] [Indexed: 12/18/2022] Open
Abstract
Mipu1 (myocardial ischemic preconditioning up-regulated protein 1), recently identified in our lab, is a novel zinc-finger transcription factor which is up-regulated during ischemic preconditioning. However, it is not clear what transcription factor contributes to its inducible expression. In the present study, we reported that HIF-1 regulates the inducible expression of Mipu1 which is involved in the cytoprotection of HIF-1α against oxidative stress by inhibiting Bax expression. Our results showed that the inducible expression of Mipu1 was associated with the expression and activation of transcription factor HIF-1 as indicated by cobalt chloride (CoCl2) treatment, HIF-1α overexpression and knockdown assays. EMSA and luciferase reporter gene assays showed that HIF-1α bound to the hypoxia response element (HRE) within Mipu1 promoter region and promoted its transcription. Moreover, our results revealed that Mipu1 inhibited the expression of Bax, an important pro-apoptosis protein associated with the intrinsic pathway of apoptosis, elevating the cytoprotection of HIF-1 against hydrogen peroxide (H2O2)-mediated injury in H9C2 cells. Our findings implied that Bax may be a potential target gene of transcription factor Mipu1, and provided a novel insight for understanding the cytoprotection of HIF-1 and new clues for further elucidating the mechanisms by which Mipu1 protects cell against pathological stress.
Collapse
Affiliation(s)
- Kangkai Wang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jian Lei
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, the Affiliated Tumor Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jiang Zou
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hui Xiao
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Anlan Chen
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiaoliu Liu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Endocrinology, the Second Xiangya hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ying Liu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Lei Jiang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zihui Xiao
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- * E-mail: (XX); (ZX)
| | - Xianzhong Xiao
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- * E-mail: (XX); (ZX)
| |
Collapse
|
42
|
Pattern of heat shock factor and heat shock protein expression in lymphocytes of bipolar patients: increased HSP70-glucocorticoid receptor heterocomplex. J Psychiatr Res 2013; 47:1725-36. [PMID: 23938235 DOI: 10.1016/j.jpsychires.2013.07.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 05/22/2013] [Accepted: 07/17/2013] [Indexed: 12/21/2022]
Abstract
Bipolar disorder (BD), a stress-related disease, is characterized by altered glucocorticoid receptor (GR) signalling. Stress response includes activation of heat shock factor (HSF) and subsequent heat shock protein (HSP) synthesis which regulate GR folding and function. The objective of this study was to investigate the possible role of HSFs, HSPs and their interaction with GR in BD. We applied immunoprecipitation, SDS-PAGE/Western blot analysis and electrophoretic mobility shift assay (EMSA) in lymphocytes (whole cell or nuclear extracts) from BD patients and healthy subjects and determined the HSPs (HSP90 and HSP70), the heterocomplexes HSP90-GR and HSP70-GR, the HSFs (HSF1 and HSF4) as well as the HSF-DNA binding. The HSP70-GR heterocomplex was elevated (p < 0.05) in BD patients vs healthy subjects, and nuclear HSP70 was reduced (p ≤ 0.01) in bipolar manic patients. Protein levels of HSF1, HSF4, HSP90, HSP90-GR heterocomplex, and HSF-DNA binding remained unaltered in BD patients vs healthy subjects. The corresponding effect sizes (ES) indicated a large ES for HSP70-GR, HSP70, HSF-DNA binding and HSF4, and a medium ES for HSP90, HSF1 and HSP90-GR between healthy subjects and bipolar patients. Significant correlations among HSFs, HSPs, GR and HSP70-GR heterocomplex were observed in healthy subjects, which were abrogated in bipolar patients. The higher interaction between GR and HSP70 and the disturbances in the relations among heat shock response parameters and GR as observed in our BD patients may provide novel insights into the contribution of these factors in BD aetiopathogenesis.
Collapse
|
43
|
IL-1β, RAGE and FABP4: targeting the dynamic trio in metabolic inflammation and related pathologies. Future Med Chem 2013; 5:1089-108. [PMID: 23795967 DOI: 10.4155/fmc.13.90] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Within the past decade, inflammatory and lipid mediators, such as IL-1β, FABP4 and RAGE, have emerged as important contributors to metabolic dysfunction. As growing experimental and clinical evidence continues to tie obesity-induced chronic inflammation with dysregulated lipid, insulin signaling and related pathologies, IL-1β, FABP4 and RAGE each are being independently implicated as culprits in these events. There are also convincing data that molecular pathways driven by these molecules are interconnected in exacerbating metabolic consequences of obesity. This article highlights the roles of IL-1β, FABP4 and RAGE in normal physiology as well as focusing specifically on their contribution to inflammation, insulin resistance, atherosclerosis, Type 2 diabetes and cancer. Studies implicating the interconnection between these pathways, current and emerging therapeutics, and their use as potential biomarkers are also discussed. Evidence of impact of IL-1β, FABP4 and RAGE pathways on severity of metabolic dysfunction underlines the strong links between inflammatory events, lipid metabolism and insulin regulation, and offers new intriguing approaches for future therapies of obesity-driven pathologies.
Collapse
|
44
|
Chen J, Zhang J, Xu L, Xu C, Chen S, Yang J, Jiang H. Inhibition of neointimal hyperplasia in the rat carotid artery injury model by a HMGB1 inhibitor. Atherosclerosis 2012; 224:332-339. [PMID: 22857898 DOI: 10.1016/j.atherosclerosis.2012.07.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 05/01/2012] [Accepted: 07/12/2012] [Indexed: 02/07/2023]
Abstract
OBJECTIVE High mobility group box 1 protein (HMGB1) is expressed in atherosclerotic lesions. However, its role in vascular system is unknown. In this study, we explore whether the inhibition of HMGB1 attenuates neointimal formation in animal models. METHODS AND RESULTS Experiments were performed with VSMCs from thoracic aorta of SD rats in vitro, and a rat carotid artery balloon injury model in vivo. HMGB1 levels were increased after stimulation of angiotensin II (Ang II) and 10% serum in cultured VSMCs. HMGB1 inhibitor (glycyrrhizin) significantly inhibited the proliferation and migration of Ang II-treated VSMCs, which was accompanied with decreased oxidative stress and inflammation. The underlying mechanisms were related with the promotion of antioxidant systems activity and deactivation of p38 MAPK/NF-κB signaling pathway, respectively. Furthermore, inhibition of HMGB1 blunted Notch signaling pathway during VSMCs phenotypic transition, and correspondingly restored VSMCs differentiated phenotype under 10% serum stimulation. In vivo study, HMGB1 expression was elevated after artery injury. Meanwhile, glycyrrhizin treatment suppressed HMGB1 expression, which was accompanied with blunted inflammation and oxidative stress after 7 days of balloon injury. Moreover, the area of neointimal to media area ratio was significantly decreased in glycyrrhizin group compared with injury group at 14 days after balloon injury. CONCLUSIONS Inhibition of HMGB1 activity attenuated VSMCs activation and neointimal formation after carotid injury. Therefore, blockage of HMGB1 might represent a novel therapeutic strategy for vascular injury.
Collapse
MESH Headings
- Angiotensin II/metabolism
- Animals
- Anti-Inflammatory Agents/pharmacology
- Antioxidants/pharmacology
- Cardiovascular Agents/pharmacology
- Carotid Arteries/drug effects
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Carotid Artery Injuries/drug therapy
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Cell Differentiation/drug effects
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Glycyrrhizic Acid/pharmacology
- HMGB1 Protein/antagonists & inhibitors
- HMGB1 Protein/metabolism
- Hyperplasia
- Inflammation Mediators/metabolism
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- NF-kappa B/metabolism
- Neointima
- Oxidative Stress/drug effects
- Phenotype
- Rats
- Rats, Sprague-Dawley
- Receptors, Notch/metabolism
- Signal Transduction/drug effects
- Time Factors
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Jing Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | | | | | | | | | | | | |
Collapse
|
45
|
Ortega E, Bote ME, Besedovsky HO, Rey AD. Hsp72, inflammation, and aging: causes, consequences, and perspectives. Ann N Y Acad Sci 2012; 1261:64-71. [DOI: 10.1111/j.1749-6632.2012.06619.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
46
|
Lee JJ, Hsiao CC, Yang IH, Chou MH, Wu CL, Wei YC, Chen CH, Chuang JH. High-mobility group box 1 protein is implicated in advanced glycation end products-induced vascular endothelial growth factor A production in the rat retinal ganglion cell line RGC-5. Mol Vis 2012; 18:838-50. [PMID: 22511847 PMCID: PMC3327441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2011] [Accepted: 04/02/2012] [Indexed: 11/09/2022] Open
Abstract
PURPOSE High-mobility group box 1 protein (HMGB1) has been reported to be a potent proangiogenic factor induced by inflammatory stress. In this study, we explore the role of HMGB1 in advanced glycation end products (AGEs)-induced vascular endothelial growth factor A (VEGF-A) production in rat retinal ganglion cell line 5 (RGC-5) cells. METHODS The VEGF-A protein and mRNA levels in conditioned medium of RGC-5 cells incubated with AGE-modified BSA (AGE-BSA) were examined with real-time PCR and enzyme-linked immunosorbent assay (ELISA), and BSA-treated cells were used as controls. The expression of HMGB1, c-Jun N-terminal kinase (JNK), extracellular-signal-regulated kinase (ERK), and p38 mitogen-activated protein kinase (p38 MAPK) was assessed with immunofluorescence and western blot analysis. Reactive oxidative species (ROS) were detected with flow cytometry measurements of peroxide-dependent oxidation of 2'-7'-dichlorofluorescein-diacetate (DCFH-DA). N-Acetyl-L-cysteine (NAC), glycyrrhizin (GZ), and SP600125 were used to block ROS, HMGB1, and JNK, respectively. RESULTS Compared with the BSA controls, the RGC-5 cells incubated with AGE-BSA showed a dose- and time-dependent increase in VEGF-A mRNA and VEGF-A protein secretion in the supernatant, with the highest levels achieved at 24 h. AGE-BSA stimulated a significant release of HMGB1 in the supernatant and a significant increase of intracellular ROS production at 3 h. NAC blocked HMGB1 production in a dose-dependent manner. Blocking with GZ, NAC, and JNK significantly suppressed AGE-induced VEGF-A production. CONCLUSIONS HMGB1 is implicated in the production of VEGF-A in retinal ganglion cell line-5 (RGC-5). Blocking HMGB1, ROS, or the JNK pathway may attenuate VEGF-A production, suggesting HMGB1 and related signaling molecules play a role in diabetic retinopathy.
Collapse
Affiliation(s)
- Jong-Jer Lee
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan,The Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chang-Chun Hsiao
- The Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - I-Hui Yang
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ming-Huei Chou
- The Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Lin Wu
- Department of Surgery, Division of Pediatric Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yin-Chu Wei
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chih-Hsin Chen
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jiin-Haur Chuang
- Department of Surgery, Division of Pediatric Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan,The Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
47
|
Yu Y, Liu M, Zhang L, Cao Q, Zhang P, Jiang H, Zou Y, Ge J. Heat shock transcription factor 1 inhibits H₂O₂-induced cardiomyocyte death through suppression of high-mobility group box 1. Mol Cell Biochem 2012; 364:263-9. [PMID: 22246807 DOI: 10.1007/s11010-012-1226-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 01/04/2012] [Indexed: 01/28/2023]
Abstract
Heat shock transcription factor 1 (HSF1), which has been identified as an endogenous cardioprotective factor, possesses considerable anti-inflammatory effects and the ability against oxidative stress. However, the mechanisms have not been fully characterized yet. In this study, we investigated the effects of HSF1-regulated HMGB1 on cardiomyocyte death. Cultured cardiomyocytes were transfected with empty vector or HSF1 plasmid before the exposure to H(2)O(2). Cell death was assessed by cell staining. Additionally, the levels of intracellular and extracellular HMGB1 as well as its subcellular location were detected, and the expression of heat shock proteins (HSP27 and HSP90) in the cardiomyocytes was also determined. Not only did H(2)O(2) significantly increase cell death, but also elevated the levels of intracellular and extracellular HMGB1 and induced its translocation, whereas, as expected, HSF1 overexpression effectively attenuated cell death. Furthermore, HSF1 inhibited the intracellular expression of HMGB1 at early stage of oxidative stress and subsequently, HSF1 did negatively regulate the extracellular levels and the translocation of HMGB1 at late stage. Besides, the expression of HSP27 and HSP90 was significantly increased. These results suggested HSF1-attenuated cardiomyocyte death via reducing intracellular and extracellular levels of HMGB1 as well as preventing its translocation, which was partially associated with HSP27 and HSP90 up-regulated by HSF1 overexpression.
Collapse
Affiliation(s)
- Ying Yu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Wang K, Deng G, Chen G, Liu M, Yi Y, Yang T, McMillan DR, Xiao X. Heat shock protein 70 inhibits hydrogen peroxide-induced nucleolar fragmentation via suppressing cleavage and down-regulation of nucleolin. Cell Stress Chaperones 2012; 17:121-30. [PMID: 21960124 PMCID: PMC3227849 DOI: 10.1007/s12192-011-0292-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 09/08/2011] [Accepted: 09/12/2011] [Indexed: 10/17/2022] Open
Abstract
It has been reported that nucleolar fragmentation is a part of the overall apoptotic morphology, however, it is currently obscure whether and how nucleolar fragmentation can be induced by hydrogen peroxide (H(2)O(2)) and heat shock protein 70 (Hsp70) can prevent nucleolar fragmentation. To dissect these two questions, C(2)C(12) myogenic cells and immortalized mouse embryonic fibroblasts (MEFs) with heat shock transcriptional factor 1 (HSF1) null mutation were treated with heat shock response (HS) (42.5 ± 0.5°C for 1 h and recovery at 37°C for 24 h) and then were insulted with 0.5 mmol/L H(2)O(2). Morphological changes of nucleoli were observed under contrast microscope or electronic microscope. It was found that (1) stimulation with H(2)O(2)-induced nucleolar fragmentation by mediating cleavage and down-regulation of nucleolar protein, nucleolin in C(2)C(12) myocytes and MEFs; (2) HS suppressed nucleolar fragmentation by inducing the expression of Hsp70 in an HSF1-dependent manner as indicated by assays of transfection with Hsp70 antisense oligonucleotides (AS-ONs) or recombinant plasmids of full-length Hsp70 cDNA; (3) protection of Hsp70 against nucleolar fragmentation was related to its accumulation in nucleolus mediated by nuclear localization sequence and its inhibition against cleavage and down-regulation of nucleolin. These results suggested that H(2)O(2)-induced nucleolar fragmentation and HS or Hsp70 inhibit H(2)O(2)-induced nucleolar fragmentation through the translocation of Hsp70 into nucleolar and its protection against impairment of nucleolin.
Collapse
Affiliation(s)
- Kangkai Wang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| | - Gonghua Deng
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| | - Guangwen Chen
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| | - Meidong Liu
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| | - Yuxin Yi
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| | - Tubao Yang
- School of Public Health, Central South University, Changsha, Hunan 410008 People’s Republic of China
| | - Daniel R. McMillan
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9063 USA
| | - Xiangzhong Xiao
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410008 People’s Republic of China
| |
Collapse
|
49
|
Huang J, Ni J, Liu K, Yu Y, Xie M, Kang R, Vernon P, Cao L, Tang D. HMGB1 promotes drug resistance in osteosarcoma. Cancer Res 2011; 72:230-8. [PMID: 22102692 DOI: 10.1158/0008-5472.can-11-2001] [Citation(s) in RCA: 209] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Osteosarcoma is the most commonly occurring bone cancer in children and adolescents. Unfortunately, treatment failures are common due to the development of chemoresistance, for which the underlying molecular mechanisms remain unclear. In this study, we implicate the DNA-binding protein HMGB1, which also exerts immunoregulatory effects in its secreted form, in the development of drug resistance in osteosarcoma. Anticancer agents including doxorubicin, cisplatin, and methotrexate each induced HMGB1 upregulation in human osteosarcoma cells, and RNA interference-mediated knockdown of HMGB1 restored the chemosensitivity of osteosarcoma cells in vivo and in vitro. Mechanistic investigation revealed that HMGB1 increased drug resistance by inducing autophagy, an intracellular self-defense mechanism known to confer drug resistance. We found that HMGB1 bound to the autophagy regulator Beclin1 and regulated the formation of the Beclin1-PI3KC3 [PI3KC3, phosphatidylinositol 3-kinase class 3] complex that facilitates autophagic progression. In addition, we found that interaction between HMGB1 and Beclin1 relied upon the autophagic complex ULK1-mAtg13-FIP200. Therefore, through its role as a regulator of autophagy, HMGB1 is a critical factor in the development of chemoresistance, and it offers a novel target for improving osteosarcoma therapy.
Collapse
Affiliation(s)
- Jun Huang
- Departments of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Programmed necrosis: backup to and competitor with apoptosis in the immune system. Nat Immunol 2011; 12:1143-9. [PMID: 22089220 DOI: 10.1038/ni.2159] [Citation(s) in RCA: 286] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Programmed cell death is essential for the development and maintenance of the immune system and its responses to exogenous and endogenous stimuli. Studies have demonstrated that in addition to caspase-dependent apoptosis, necrosis dependent on the kinases RIP1 and RIP3 (also called necroptosis) is a major programmed cell-death pathway in development and immunity. These two programmed cell-death pathways may suppress each other, and necroptosis also serves as an alternative when caspase-dependent apoptosis is inhibited or absent. Here we summarize recent advancements that have identified the molecular mechanisms that underlie necroptosis and explore the mechanisms that regulate the interplay between apoptosis and necroptosis.
Collapse
|