1
|
Li Y, Li D, Jiang Z, Yuan Z, Sun Z, Sun L. D-M159 Synergistically Induces Apoptosis in HeLa Cells Through Endoplasmic Reticulum Stress and Mitochondrial Dysfunction. Int J Mol Sci 2025; 26:3172. [PMID: 40243937 DOI: 10.3390/ijms26073172] [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: 03/01/2025] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Pore-forming peptides are promising antimicrobial and anticancer agents due to their membrane selectivity and biodegradability. Our prior work identified peptide M159, which permeabilized synthetic phosphatidylcholine liposomes without mammalian cell toxicity. Here, we report that the D-type variant (D-M159) induces apoptosis in HeLa cells under starvation. To explore its anticancer mechanism, we analyzed D-M159 cytotoxicity, intracellular uptake, and apoptotic pathways via flow cytometry, confocal microscopy, and Western blot. Calcium dynamics and mitochondrial function were examined via specific labeling and functional assays. Results revealed that D-M159 exhibited starvation-dependent, dose-responsive cytotoxicity and triggered apoptosis in HeLa cells. Mechanistic studies indicated that D-M159 entered the cells via caveolin-dependent and caveolae-dependent endocytosis pathways and induced endoplasmic reticulum stress in HeLa cells by up-regulating proteins such as ATF6, p-IRE1, PERK, GRP78, and CHOP. Meanwhile, D-M159 promoted the expression of IP3R1, GRP75, and VDAC1, which led to mitochondrial calcium iron overload, decreased mitochondrial membrane potential, and increased reactive oxygen species (ROS) generation, thereby activating the mitochondrial apoptotic pathway and inducing the aberrant expression of Bax, Bcl-2, Caspase-9, and Caspase-3. This study showed that D-M159 synergistically induced apoptosis in starved HeLa cells through endoplasmic reticulum stress and mitochondrial dysfunction, demonstrating its potential as a novel anticancer agent.
Collapse
Affiliation(s)
- Yuanyuan Li
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Pharmacy, Hunan Normal University, Changsha 410013, China
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Dingding Li
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Pharmacy, Hunan Normal University, Changsha 410013, China
| | - Zonghan Jiang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Zhihang Yuan
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Zhiliang Sun
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Leisheng Sun
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Pharmacy, Hunan Normal University, Changsha 410013, China
| |
Collapse
|
2
|
Chang LC, Yin MT, Laird GM, Ritter KD, Shah JG, Debnath AK. A First-in-Class Dual Degrader of Bcl-2/Bcl-xL Reverses HIV Latency and Minimizes Ex Vivo Reservoirs from Patients. Int J Mol Sci 2025; 26:2772. [PMID: 40141414 PMCID: PMC11942780 DOI: 10.3390/ijms26062772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
The persistence of latent HIV-1 proviruses in CD4+ T cells is a major obstacle to curing HIV. The "shock and kill" strategy involves reversing latency with latency-reversing agents (LRAs) and selectively inducing cell death in infected cells. However, current LRAs have shown limited efficacy in eliminating the ex vivo HIV reservoir and thus failed in clinical study. In this study, we repurposed PZ703b, a pro-apoptotic protein degrader initially developed for anti-leukemia therapy, to target HIV eradication. PZ703b induced the degradation of Bcl-2 and Bcl-xL, activating the non-canonical NF-kB pathway and caspases cascade, resulting in latency reversal and the selective apoptosis of infected cells. The treatment of ex vivo CD4+ T cells from ART-suppressed HIV-1 patients led to approximately a 50% reduction in the replication-competent reservoir. While this result does not reach the threshold required for a complete cure, it demonstrates the potential of a dual degrader of Bcl-2/Bcl-xL in reversing HIV latency and inducing selective cell death. Our study provides a proof-of-concept for using dual degraders of Bcl-2/Bcl-xL as a novel category of LRAs in therapeutic strategies aimed at reducing HIV reservoirs. This approach may pave the way for the further exploration of targeted interventions to eliminate the HIV-inducible reservoir.
Collapse
Affiliation(s)
- Lin-Chun Chang
- Laboratory of Molecular Modeling and Drug Design, Lindsey F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| | - Michael T. Yin
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.T.Y.)
| | - Gregory M. Laird
- Accelevir Diagnostics, Baltimore, MD 21202, USA; (G.M.L.); (K.D.R.)
| | | | - Jayesh G. Shah
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.T.Y.)
| | - Asim K. Debnath
- Laboratory of Molecular Modeling and Drug Design, Lindsey F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| |
Collapse
|
3
|
Mivehchi H, Eskandari-Yaghbastlo A, Pour Bahrami P, Elhami A, Faghihinia F, Nejati ST, Kazemi KS, Nabi Afjadi M. Exploring the role of oral bacteria in oral cancer: a narrative review. Discov Oncol 2025; 16:242. [PMID: 40009328 DOI: 10.1007/s12672-025-01998-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/19/2025] [Indexed: 02/27/2025] Open
Abstract
A growing body of research indicates that a wide range of cancer types may correlate with human microbiome components. On the other hand, little is known about the potential contribution of the oral microbiota to oral cancer. However, some oral microbiome components can stimulate different tumorigenic processes associated with the development of cancer. In this line, two prevalent oral infections, Porphyromonas gingivalis, and Fusobacterium nucleatum can increase tumor growth. The microbiome can impact the course of the illness through direct interactions with the human body and major modifications to the toxicity and responsiveness to different kinds of cancer therapy. Recent research has demonstrated a relationship between specific phylogenetic groupings and the results of immunotherapy treatment for particular tumor types. Conversely, there has been a recent upsurge in interest in the possibility of using microbes to treat cancer. At the moment, some species, such as Salmonella typhimurium and Clostridium spp., are being explored as possible cancer treatment vectors. Thus, understanding these microbial interactions highlights the importance of maintaining a healthy oral microbiome in preventing oral cancers. From this perspective, this review will discuss the role of the microbiome on oral cancers and their possible application in oral cancer treatment/improvement.
Collapse
Affiliation(s)
- Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | | | | | - Anis Elhami
- Faculty of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farbod Faghihinia
- School of Dentistry, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Kimia Sadat Kazemi
- Faculty of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
4
|
Gheorghe SR, Crăciun AM, Ilyés T, Tisa IB, Sur L, Lupan I, Samasca G, Silaghi CN. Converging Mechanisms of Vascular and Cartilaginous Calcification. BIOLOGY 2024; 13:565. [PMID: 39194503 DOI: 10.3390/biology13080565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Physiological calcification occurs in bones and epiphyseal cartilage as they grow, whereas ectopic calcification occurs in blood vessels, cartilage, and soft tissues. Although it was formerly thought to be a passive and degenerative process associated with aging, ectopic calcification has been identified as an active cell-mediated process resembling osteogenesis, and an increasing number of studies have provided evidence for this paradigm shift. A significant association between vascular calcification and cardiovascular risk has been demonstrated by various studies, which have shown that arterial calcification has predictive value for future coronary events. With respect to cartilaginous calcification, calcium phosphate or hydroxyapatite crystals can form asymptomatic deposits in joints or periarticular tissues, contributing to the pathophysiology of osteoarthritis, rheumatoid arthritis, ankylosing spondylitis, tendinitis, and bursitis. The risk factors and sequence of events that initiate ectopic calcification, as well as the mechanisms that prevent the development of this pathology, are still topics of debate. Consequently, in this review, we focus on the nexus of the mechanisms underlying vascular and cartilaginous calcifications, trying to circumscribe the similarities and disparities between them to provide more clarity in this regard.
Collapse
Affiliation(s)
- Simona R Gheorghe
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alexandra M Crăciun
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Tamás Ilyés
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ioana Badiu Tisa
- Department of Pediatrics III, Iuliu Hatieganu University of Medicine and Pharmacy, 400217 Cluj-Napoca, Romania
| | - Lucia Sur
- Department of Pediatrics I, Iuliu Hatieganu University of Medicine and Pharmacy, 400370 Cluj-Napoca, Romania
| | - Iulia Lupan
- Department of Molecular Biology, Babes-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Gabriel Samasca
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Ciprian N Silaghi
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
5
|
Petrache I, Riches DW. Reduced Bik expression drives low-grade airway inflammation and increased risk for COPD in females. J Clin Invest 2024; 134:e177753. [PMID: 38357926 PMCID: PMC10866644 DOI: 10.1172/jci177753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Abstract
Chronic low-grade inflammation is increasingly recognized as a subtle yet potent risk factor for a multitude of age-related disorders, including respiratory diseases, cardiovascular conditions, metabolic syndromes, autoimmunity, and cancer. In this issue of the JCI, Mebratu, Jones, and colleagues shed new light on the mechanisms that promote low-grade airway inflammation and how this contributes to the development of chronic obstructive pulmonary disease (COPD). Their finding that Bik deficiency leads to spontaneous emphysema in female mice, but not in males, marks a notable advancement in our understanding of how inflammatory processes can diverge based on biological sex. This finding is of clinical relevance, given the vulnerability of women to developing COPD.
Collapse
Affiliation(s)
- Irina Petrache
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - David W.H. Riches
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
- Department of Research, Veterans Affairs Eastern Colorado Health Care System, Denver, Colorado, USA
| |
Collapse
|
6
|
Pollenus E, Possemiers H, Knoops S, Prenen F, Vandermosten L, Thienpont C, Abdurahiman S, Demeyer S, Cools J, Matteoli G, Vanoirbeek JAJ, Vande Velde G, Van den Steen PE. Single cell RNA sequencing reveals endothelial cell killing and resolution pathways in experimental malaria-associated acute respiratory distress syndrome. PLoS Pathog 2024; 20:e1011929. [PMID: 38236930 PMCID: PMC10826972 DOI: 10.1371/journal.ppat.1011929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/30/2024] [Accepted: 12/29/2023] [Indexed: 01/31/2024] Open
Abstract
Plasmodium parasites cause malaria, a global health disease that is responsible for more than 200 million clinical cases and 600 000 deaths each year. Most deaths are caused by various complications, including malaria-associated acute respiratory distress syndrome (MA-ARDS). Despite the very rapid and efficient killing of parasites with antimalarial drugs, 15% of patients with complicated malaria succumb. This stresses the importance of investigating resolution mechanisms that are involved in the recovery from these complications once the parasite is killed. To study the resolution of MA-ARDS, P. berghei NK65-infected C57BL/6 mice were treated with antimalarial drugs after onset of symptoms, resulting in 80% survival. Micro-computed tomography revealed alterations of the lungs upon infection, with an increase in total and non-aerated lung volume due to edema. Whole body plethysmography confirmed a drastically altered lung ventilation, which was restored during resolution. Single-cell RNA sequencing indicated an increased inflammatory state in the lungs upon infection, which was accompanied by a drastic decrease in endothelial cells, consistent with CD8+ T cell-mediated killing. During resolution, anti-inflammatory pathways were upregulated and proliferation of endothelial cells was observed. MultiNicheNet interactome analysis identified important changes in the ligand-receptor interactions during disease resolution that warrant further exploration in order to develop new therapeutic strategies. In conclusion, our study provides insights in pro-resolving pathways that limit inflammation and promote endothelial cell proliferation in experimental MA-ARDS. This information may be useful for the design of adjunctive treatments to enhance resolution after Plasmodium parasite killing by antimalarial drugs.
Collapse
Affiliation(s)
- Emilie Pollenus
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Hendrik Possemiers
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Sofie Knoops
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Fran Prenen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Leen Vandermosten
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Chloë Thienpont
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Saeed Abdurahiman
- Laboratory of Mucosal Immunology, Translational Research in Gastro-Intestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Sofie Demeyer
- Laboratory of Molecular Biology of Leukemia, Department of Human Genetics, VIB—KU Leuven, Leuven, Belgium
| | - Jan Cools
- Laboratory of Molecular Biology of Leukemia, Department of Human Genetics, VIB—KU Leuven, Leuven, Belgium
| | - Gianluca Matteoli
- Laboratory of Mucosal Immunology, Translational Research in Gastro-Intestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Jeroen A. J. Vanoirbeek
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Greetje Vande Velde
- Biomedical MRI, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Philippe E. Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Mebratu YA, Jones JT, Liu C, Negasi ZH, Rahman M, Rojas-Quintero J, O’Connor GT, Gao W, Dupuis J, Cho MH, Litonjua AA, Randell S, Tesfaigzi Y. Bik promotes proteasomal degradation to control low-grade inflammation. J Clin Invest 2023; 134:e170594. [PMID: 38113109 PMCID: PMC10866658 DOI: 10.1172/jci170594] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
Although chronic low-grade inflammation does not cause immediate clinical symptoms, over the longer term, it can enhance other insults or age-dependent damage to organ systems and thereby contribute to age-related disorders, such as respiratory disorders, heart disease, metabolic disorders, autoimmunity, and cancer. However, the molecular mechanisms governing low-level inflammation are largely unknown. We discovered that Bcl-2-interacting killer (Bik) deficiency causes low-level inflammation even at baseline and the development of spontaneous emphysema in female but not male mice. Similarly, a single nucleotide polymorphism that reduced Bik levels was associated with increased inflammation and enhanced decline in lung function in humans. Transgenic expression of Bik in the airways of Bik-deficient mice inhibited allergen- or LPS-induced lung inflammation and reversed emphysema in female mice. Bik deficiency increased nuclear but not cytosolic p65 levels because Bik, by modifying the BH4 domain of Bcl-2, interacted with regulatory particle non-ATPase 1 (RPN1) and RPN2 and enhanced proteasomal degradation of nuclear proteins. Bik deficiency increased inflammation primarily in females because Bcl-2 and Bik levels were reduced in lung tissues and airway cells of female compared with male mice. Therefore, controlling low-grade inflammation by modifying the unappreciated role of Bik and Bcl-2 in facilitating proteasomal degradation of nuclear proteins may be crucial in treating chronic age-related diseases.
Collapse
Affiliation(s)
- Yohannes A. Mebratu
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Jane T. Jones
- University Geisel School of Medicine, Department of Microbiology and Immunology, Dartmouth, Hanover, New Hampshire, USA
| | - Congjian Liu
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Zerihun H. Negasi
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Mizanur Rahman
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Joselyn Rojas-Quintero
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - George T. O’Connor
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- National Heart, Lung, and Blood Institute’s (NHLBI’s) Framingham Heart Study, Framingham, Massachusetts, USA
| | - Wei Gao
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
- Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Scott Randell
- Marsico Lung Institute, UNC School of Medicine, Chapel Hill, North Carolina, USA
| | - Yohannes Tesfaigzi
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Riviere-Cazaux C, Carlstrom LP, Neth BJ, Olson IE, Rajani K, Rahman M, Ikram S, Mansour MA, Mukherjee B, Warrington AE, Short SC, von Zglinicki T, Brown DA, Burma S, Tchkonia T, Schafer MJ, Baker DJ, Kizilbash SH, Kirkland JL, Burns TC. An untapped window of opportunity for glioma: targeting therapy-induced senescence prior to recurrence. NPJ Precis Oncol 2023; 7:126. [PMID: 38030881 PMCID: PMC10687268 DOI: 10.1038/s41698-023-00476-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
High-grade gliomas are primary brain tumors that are incredibly refractory long-term to surgery and chemoradiation, with no proven durable salvage therapies for patients that have failed conventional treatments. Post-treatment, the latent glioma and its microenvironment are characterized by a senescent-like state of mitotic arrest and a senescence-associated secretory phenotype (SASP) induced by prior chemoradiation. Although senescence was once thought to be irreversible, recent evidence has demonstrated that cells may escape this state and re-enter the cell cycle, contributing to tumor recurrence. Moreover, senescent tumor cells could spur the growth of their non-senescent counterparts, thereby accelerating recurrence. In this review, we highlight emerging evidence supporting the use of senolytic agents to ablate latent, senescent-like cells that could contribute to tumor recurrence. We also discuss how senescent cell clearance can decrease the SASP within the tumor microenvironment thereby reducing tumor aggressiveness at recurrence. Finally, senolytics could improve the long-term sequelae of prior therapy on cognition and bone marrow function. We critically review the senolytic drugs currently under preclinical and clinical investigation and the potential challenges that may be associated with deploying senolytics against latent glioma. In conclusion, senescence in glioma and the microenvironment are critical and potential targets for delaying or preventing tumor recurrence and improving patient functional outcomes through senotherapeutics.
Collapse
Affiliation(s)
| | | | | | - Ian E Olson
- Department of Neurological Surgery, Northwestern University, Chicago, IL, USA
| | | | - Masum Rahman
- Department of Neurological Surgery, Rochester, MN, USA
| | - Samar Ikram
- Department of Neurological Surgery, Rochester, MN, USA
| | | | - Bipasha Mukherjee
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Arthur E Warrington
- Department of Neurological Surgery, Rochester, MN, USA
- Department of Neurology, Rochester, MN, USA
| | - Susan C Short
- Leeds Institute of Medical Research at St. James's, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Thomas von Zglinicki
- Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Desmond A Brown
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Sandeep Burma
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Tamar Tchkonia
- Department of Physiology and Biomedical Engineering, Rochester, MN, USA
| | - Marissa J Schafer
- Department of Physiology and Biomedical Engineering, Rochester, MN, USA
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Rochester, MN, USA
| | | | - James L Kirkland
- Department of Pediatric and Adolescent Medicine, Rochester, MN, USA
- Department of Medicine, Rochester, MN, USA
| | - Terry C Burns
- Department of Neurological Surgery, Rochester, MN, USA.
| |
Collapse
|
9
|
Al-Sawaf O, Zhang C, Jin HY, Robrecht S, Choi Y, Balasubramanian S, Kotak A, Chang YM, Fink AM, Tausch E, Schneider C, Ritgen M, Kreuzer KA, Chyla B, Paulson JN, Pallasch CP, Frenzel LP, Peifer M, Eichhorst B, Stilgenbauer S, Jiang Y, Hallek M, Fischer K. Transcriptomic profiles and 5-year results from the randomized CLL14 study of venetoclax plus obinutuzumab versus chlorambucil plus obinutuzumab in chronic lymphocytic leukemia. Nat Commun 2023; 14:2147. [PMID: 37072421 PMCID: PMC10113251 DOI: 10.1038/s41467-023-37648-w] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 03/24/2023] [Indexed: 04/20/2023] Open
Abstract
Data on long-term outcomes and biological drivers associated with depth of remission after BCL2 inhibition by venetoclax in the treatment of chronic lymphocytic leukemia (CLL) are limited. In this open-label parallel-group phase-3 study, 432 patients with previously untreated CLL were randomized (1:1) to receive either 1-year venetoclax-obinutuzumab (Ven-Obi, 216 patients) or chlorambucil-Obi (Clb-Obi, 216 patients) therapy (NCT02242942). The primary endpoint was investigator-assessed progression-free survival (PFS); secondary endpoints included minimal residual disease (MRD) and overall survival. RNA sequencing of CD19-enriched blood was conducted for exploratory post-hoc analyses. After a median follow-up of 65.4 months, PFS is significantly superior for Ven-Obi compared to Clb-Obi (Hazard ratio [HR] 0.35 [95% CI 0.26-0.46], p < 0.0001). At 5 years after randomization, the estimated PFS rate is 62.6% after Ven-Obi and 27.0% after Clb-Obi. In both arms, MRD status at the end of therapy is associated with longer PFS. MRD + ( ≥ 10-4) status is associated with increased expression of multi-drug resistance gene ABCB1 (MDR1), whereas MRD6 (< 10-6) is associated with BCL2L11 (BIM) expression. Inflammatory response pathways are enriched in MRD+ patient solely in the Ven-Obi arm. These data indicate sustained long-term efficacy of fixed-duration Ven-Obi in patients with previously untreated CLL. The distinct transcriptomic profile of MRD+ status suggests possible biological vulnerabilities.
Collapse
Affiliation(s)
- Othman Al-Sawaf
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany.
- Cancer Institute, University College London, London, UK.
- Francis Crick Institute, London, UK.
| | - Can Zhang
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
| | | | - Sandra Robrecht
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
| | - Yoonha Choi
- Genentech Inc., South San Francisco, CA, USA
| | | | - Alex Kotak
- Roche Products Ltd, Welwyn Garden City, UK
| | | | - Anna Maria Fink
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
| | - Eugen Tausch
- Department III of Internal Medicine, Ulm University, Ulm, Germany
| | | | - Matthias Ritgen
- Department II of Internal Medicine, University of Schleswig Holstein, Kiel, Germany
| | - Karl-Anton Kreuzer
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
| | | | | | - Christian P Pallasch
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
| | - Lukas P Frenzel
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
| | - Martin Peifer
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany
| | - Barbara Eichhorst
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
| | | | | | - Michael Hallek
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany.
| | - Kirsten Fischer
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany.
| |
Collapse
|
10
|
Rogers NM, Zammit N, Nguyen-Ngo D, Souilmi Y, Minhas N, Meijles DN, Self E, Walters SN, Warren J, Cultrone D, El-Rashid M, Li J, Chtanova T, O'Connell PJ, Grey ST. The impact of the cytoplasmic ubiquitin ligase TNFAIP3 gene variation on transcription factor NF-κB activation in acute kidney injury. Kidney Int 2023; 103:1105-1119. [PMID: 37097268 DOI: 10.1016/j.kint.2023.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 02/08/2023] [Accepted: 02/23/2023] [Indexed: 04/26/2023]
Abstract
Nuclear factor κB (NF-κB) activation is a deleterious molecular mechanism that drives acute kidney injury (AKI) and manifests in transplanted kidneys as delayed graft function. The TNFAIP3 gene encodes A20, a cytoplasmic ubiquitin ligase and a master negative regulator of the NF- κB signaling pathway. Common population-specific TNFAIP3 coding variants that reduce A20's enzyme function and increase NF- κB activation have been linked to heightened protective immunity and autoimmune disease, but have not been investigated in AKI. Here, we functionally identified a series of unique human TNFAIP3 coding variants linked to the autoimmune genome-wide association studies single nucleotide polymorphisms of F127C; namely F127C;R22Q, F127C;G281E, F127C;W448C and F127C;N449K that reduce A20's anti-inflammatory function in an NF- κB reporter assay. To investigate the impact of TNFAIP3 hypomorphic coding variants in AKI we tested a mouse Tnfaip3 hypomorph in a model of ischemia reperfusion injury (IRI). The mouse Tnfaip3 coding variant I325N increases NF- κB activation without overt inflammatory disease, providing an immune boost as I325N mice exhibit enhanced innate immunity to a bacterial challenge. Surprisingly, despite exhibiting increased intra-kidney NF- κB activation with inflammation in IRI, the kidney of I325N mice was protected. The I325N variant influenced the outcome of IRI by changing the dynamic expression of multiple cytoprotective mechanisms, particularly by increasing NF- κB-dependent anti-apoptotic factors BCL-2, BCL-XL, c-FLIP and A20, altering the active redox state of the kidney with a reduction of superoxide levels and the enzyme super oxide dismutase-1, and enhancing cellular protective mechanisms including increased Foxp3+ T cells. Thus, TNFAIP3 gene variants represent a kidney and population-specific molecular factor that can dictate the course of IRI.
Collapse
Affiliation(s)
- Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia; Renal and Transplant Medicine Unit, Westmead Hospital, Westmead, New South Wales, Australia; Westmead Clinical School, University of Sydney, New South Wales, Australia
| | - Nathan Zammit
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Danny Nguyen-Ngo
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Yassine Souilmi
- Australian Centre for Ancient DNA, School of Biological Sciences, University of Adelaide, South Australia, Australia; Environment Institute, Faculty of Sciences, University of Adelaide, South Australia, Australia
| | - Nikita Minhas
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Daniel N Meijles
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Eleanor Self
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Stacey N Walters
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Joanna Warren
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Daniele Cultrone
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Maryam El-Rashid
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Jennifer Li
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Tatyana Chtanova
- Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Innate and Tumour Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Philip J O'Connell
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia; Renal and Transplant Medicine Unit, Westmead Hospital, Westmead, New South Wales, Australia; Westmead Clinical School, University of Sydney, New South Wales, Australia
| | - Shane T Grey
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
11
|
Zhao B, Li B, Zhang J, Qi Y, Chen B, Chen L. Construction of miRNA-mRNA Regulatory Network to Identify Potential Biomarkers in Infantile Hemangioma by Integrated Bioinformatics Analysis. Crit Rev Eukaryot Gene Expr 2023; 33:61-71. [PMID: 37199314 DOI: 10.1615/critreveukaryotgeneexpr.v33.i5.60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Infantile hemangioma (IH) is the most common vascular tumor among infants and children. However, the understanding of pathogenesis about IH has not been fully elucidated, and the potential diagnostic maker remains further explored. In this study, we aimed to find miRNAs as potential biomarkers of IH through bioinformatic analysis. The microarray datasets GSE69136, GSE100682 were downloaded from the GEO database. The co-expressed differential miRNAs were identified by analyzing these two datasets. The downstream common target genes were predicted by the ENCORI, Mirgene, miRWalk, and Targetscan databases. GO annotation and KEGG pathway enrichment analysis for target genes were performed. The STRING database and Cytoscape software were used to construct the protein-protein interaction network and screen hub genes. Then potential diagnostic markers for IH were further screened and identified by using Receiver operating characteristic curve analysis. A total of thirteen co-expressed up-regulated miRNAs were screened out in the above two datasets, and 778 down-regulated target genes were then predicted. GO annotation and KEGG pathway enrichment analysis indicated that the common target genes strongly correlated with IH. Through the DEM-hub gene network construction, six miRNAs associated with the hub genes were identified. Finally, has-miR-522-3p, has-miR-512-3p, has-miR-520a-5p with high diagnostic values were screened out by receiver operating characteristic analysis. In the study, the potential miRNA-mRNA regulatory network was firstly constructed in IH. And, the three miRNAs might be used as potential biomarkers for IH, which also provided novel strategies for the therapeutic intervention of IH.
Collapse
Affiliation(s)
- Boming Zhao
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Bin Li
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Jun Zhang
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Yongjian Qi
- Department of Spine Surgery and Musculoskeletal Tumor, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Biao Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| |
Collapse
|
12
|
Li Y, Faiz A, Moshage H, Schilling L, Kamps JAAM. Responses of retinal and brain microvasculature to streptozotocin induced diabetes revealed by global expression profiling. Diab Vasc Dis Res 2023; 20:14791641221147533. [PMID: 36606460 PMCID: PMC9982389 DOI: 10.1177/14791641221147533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
This study aims to determine the effects of diabetes in the retinal and brain microvasculature through gene expression profiling. Twelve male Wistar rats were randomly divided into two groups: streptozotocin-induced diabetic rats and time-matched nondiabetic rats. The retinal microvessels (RMVs) and brain microvessels (BMVs) were mechanically isolated from individual rats. Differentially expressed genes (DEGs) in diabetic and nondiabetic microvessels were identified by cDNA microarrays analysis. In RMVs, we identified 43 DEGs, of which 20 were upregulated while 23 were downregulated by diabetes. In BMVs, 35 genes DEGs were identified, of which 22 were upregulated and 13 were downregulated by diabetes. Altered expression of the Nars, Gars, Mars, Iars, Yars, Bcl2, Nqo1, NR4A3, Gpd1, Stc1, Tsc22d3, Tnfrsf21 mRNA as observed in the microarray analyses, was confirmed by quantitative RT-PCR. The aminoacyl-tRNA synthetases (aaRSs) pathway in RMVs was significantly overrepresented as compared to BMVs. Our study demonstrates for the first time that in the brain microvasculature multiple compensatory mechanisms exists, serving to protect brain tissue from diabetic insults, whereas these mechanisms are not activated in the retinal microvasculature. This provides new insights as to why brain microvasculature is less susceptible to diabetes.
Collapse
Affiliation(s)
- Youhai Li
- Division of Neurosurgical Research, Heidelberg University, Mannheim, Germany; European Center of Angioscience, Medical
Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Pathology and Medical
Biology, University Medical Center
Groningen, Groningen, The Netherlands
| | - Alen Faiz
- Department of Pathology and Medical
Biology, University Medical Center
Groningen, Groningen, The Netherlands
| | - Han Moshage
- Department of Gastroenterology and
Hepatology, University Medical Center
Groningen, Groningen, The Netherlands
| | - Lothar Schilling
- Division of Neurosurgical Research, Heidelberg University, Mannheim, Germany; European Center of Angioscience, Medical
Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jan AAM Kamps
- Department of Pathology and Medical
Biology, University Medical Center
Groningen, Groningen, The Netherlands
- Jan AAM Kamps, Department of Pathology and
Medical Biology, University of Groningen, University Medical Center Groningen,
Hanzeplein 1 (EA11), 9713GZ Groningen, The Netherlands.
| |
Collapse
|
13
|
Neuroprotective Effect of Bcl-2 on Lipopolysaccharide-Induced Neuroinflammation in Cortical Neural Stem Cells. Int J Mol Sci 2022; 23:ijms23126399. [PMID: 35742844 PMCID: PMC9223771 DOI: 10.3390/ijms23126399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 11/17/2022] Open
Abstract
Neuroinflammation is involved in the pathogenesis of neurodegenerative diseases due to increased levels of pro-inflammatory cytokines in the central nervous system (CNS). Chronic neuroinflammation induced by neurotoxic molecules accelerates neuronal damage. B-cell lymphoma 2 (Bcl-2) is generally accepted to be an important anti-apoptotic factor. However, the role of Bcl-2 in neuroprotection against neuroinflammation remains to be determined. The purpose of this study was to investigate the neuroprotective effect of Bcl-2 on lipopolysaccharide (LPS)-induced neuroinflammation in cortical neural stem cells (NSCs). LPS decreased mRNA and protein levels of Tuj-1, a neuron marker, and also suppressed neurite outgrowth, indicating that LPS results in inhibition of neuronal differentiation of NSCs. Furthermore, LPS treatment inhibited Bcl-2 expression during neuronal differentiation; inhibition of neuronal differentiation by LPS was rescued by Bcl-2 overexpression. LPS-induced pro-inflammatory cytokines, including interleukin (IL)-6 and tumor necrosis factor alpha (TNF-α), were decreased by Bcl-2 overexpression. Conversely, Bcl-2 siRNA increased the LPS-induced levels of IL-6 and TNF-α, and decreased neuronal differentiation of NSCs, raising the possibility that Bcl-2 mediates neuronal differentiation by inhibiting the LPS-induced inflammatory response in NSC. These results suggest that Bcl-2 has a neuroprotective effect by inhibiting the LPS-induced inflammatory response in NSCs.
Collapse
|
14
|
Sora V, Papaleo E. Structural Details of BH3 Motifs and BH3-Mediated Interactions: an Updated Perspective. Front Mol Biosci 2022; 9:864874. [PMID: 35685242 PMCID: PMC9171138 DOI: 10.3389/fmolb.2022.864874] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/13/2022] [Indexed: 11/18/2022] Open
Abstract
Apoptosis is a mechanism of programmed cell death crucial in organism development, maintenance of tissue homeostasis, and several pathogenic processes. The B cell lymphoma 2 (BCL2) protein family lies at the core of the apoptotic process, and the delicate balance between its pro- and anti-apoptotic members ultimately decides the cell fate. BCL2 proteins can bind with each other and several other biological partners through the BCL2 homology domain 3 (BH3), which has been also classified as a possible Short Linear Motif and whose distinctive features remain elusive even after decades of studies. Here, we aim to provide an updated overview of the structural features characterizing BH3s and BH3-mediated interactions (with a focus on human proteins), elaborating on the plasticity of BCL2 proteins and the motif properties. We also discussed the implication of these findings for the discovery of interactors of the BH3-binding groove of BCL2 proteins and the design of mimetics for therapeutic purposes.
Collapse
Affiliation(s)
- Valentina Sora
- Cancer Structural Biology, Danish Cancer Society Research Center, Copenhagen, Denmark
- Cancer Systems Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Elena Papaleo
- Cancer Structural Biology, Danish Cancer Society Research Center, Copenhagen, Denmark
- Cancer Systems Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- *Correspondence: Elena Papaleo, ,
| |
Collapse
|
15
|
Oultram JMJ, Pegler JL, Bowser TA, Ney LJ, Eamens AL, Grof CPL. Cannabis sativa: Interdisciplinary Strategies and Avenues for Medical and Commercial Progression Outside of CBD and THC. Biomedicines 2021; 9:biomedicines9030234. [PMID: 33652704 PMCID: PMC7996784 DOI: 10.3390/biomedicines9030234] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/16/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Cannabis sativa (Cannabis) is one of the world’s most well-known, yet maligned plant species. However, significant recent research is starting to unveil the potential of Cannabis to produce secondary compounds that may offer a suite of medical benefits, elevating this unique plant species from its illicit narcotic status into a genuine biopharmaceutical. This review summarises the lengthy history of Cannabis and details the molecular pathways that underpin the production of key secondary metabolites that may confer medical efficacy. We also provide an up-to-date summary of the molecular targets and potential of the relatively unknown minor compounds offered by the Cannabis plant. Furthermore, we detail the recent advances in plant science, as well as synthetic biology, and the pharmacology surrounding Cannabis. Given the relative infancy of Cannabis research, we go on to highlight the parallels to previous research conducted in another medically relevant and versatile plant, Papaver somniferum (opium poppy), as an indicator of the possible future direction of Cannabis plant biology. Overall, this review highlights the future directions of cannabis research outside of the medical biology aspects of its well-characterised constituents and explores additional avenues for the potential improvement of the medical potential of the Cannabis plant.
Collapse
Affiliation(s)
- Jackson M. J. Oultram
- Centre for Plant Science, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; (J.M.J.O.); (J.L.P.); (A.L.E.)
| | - Joseph L. Pegler
- Centre for Plant Science, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; (J.M.J.O.); (J.L.P.); (A.L.E.)
| | - Timothy A. Bowser
- CannaPacific Pty Ltd., 109 Ocean Street, Dudley, NSW 2290, Australia;
| | - Luke J. Ney
- School of Psychological Sciences, University of Tasmania, Hobart, TAS 7005, Australia;
| | - Andrew L. Eamens
- Centre for Plant Science, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; (J.M.J.O.); (J.L.P.); (A.L.E.)
| | - Christopher P. L. Grof
- Centre for Plant Science, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; (J.M.J.O.); (J.L.P.); (A.L.E.)
- CannaPacific Pty Ltd., 109 Ocean Street, Dudley, NSW 2290, Australia;
- Correspondence: ; Tel.: +612-4921-5858
| |
Collapse
|
16
|
Boraldi F, Lofaro FD, Quaglino D. Apoptosis in the Extraosseous Calcification Process. Cells 2021; 10:cells10010131. [PMID: 33445441 PMCID: PMC7827519 DOI: 10.3390/cells10010131] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 12/13/2022] Open
Abstract
Extraosseous calcification is a pathologic mineralization process occurring in soft connective tissues (e.g., skin, vessels, tendons, and cartilage). It can take place on a genetic basis or as a consequence of acquired chronic diseases. In this last case, the etiology is multifactorial, including both extra- and intracellular mechanisms, such as the formation of membrane vesicles (e.g., matrix vesicles and apoptotic bodies), mitochondrial alterations, and oxidative stress. This review is an overview of extraosseous calcification mechanisms focusing on the relationships between apoptosis and mineralization in cartilage and vascular tissues, as these are the two tissues mostly affected by a number of age-related diseases having a progressively increased impact in Western Countries.
Collapse
Affiliation(s)
- Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.D.L.); (D.Q.)
- Correspondence:
| | - Francesco Demetrio Lofaro
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.D.L.); (D.Q.)
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.D.L.); (D.Q.)
- Interuniversity Consortium for Biotechnologies (CIB), Italy
| |
Collapse
|
17
|
Long non-coding RNA maternally expressed gene regulates cigarette smoke extract induced lung inflammation and human bronchial epithelial apoptosis via miR-149-3p. Exp Ther Med 2020; 21:60. [PMID: 33365060 PMCID: PMC7716647 DOI: 10.3892/etm.2020.9492] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 07/29/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) has become a significant public health risk. Long non-coding RNAs (lncRNAs) have been identified as important factors involved in the proliferation, apoptosis and inflammatory cytokine expression of lung cells. Peripheral blood samples from 66 subjects (18 non-smokers, 24 smokers without COPD and 28 smokers with COPD) and HBE135-E6E7 cell treated with cigarette smoke extract (CSE) or not were used as the research object. The aim of the present study was to investigate the underlying mechanism of lncRNA maternally expressed gene 3 (MEG3) in COPD. Following transfection with microRNA (miR)-149-3p mimics, miR-negative control mimics, miR-149-3p inhibitor, miR-negative control inhibitor, small interfering (si)RNA targeting MEG3 (si-MEG3) and si-negative control (si-NC), levels of MEG3 and microRNA (miR)-149-3p were detected using reverse transcription-quantitative PCR, Proliferation and apoptosis were examined using the Cell Counting Kit-8 and flow cytometry assays, respectively. Enzyme-linked immunosorbent assay (ELISA) was performed to detect the expression of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). Protein levels of B-cell lymphoma-2 (Bcl-2), cleaved-caspase-3, cleaved-caspase-9, phosphorylated (p)-p65, total (t)-p65, p-lkBα and t-lkBα were measured by western blotting. Luciferase assay was conducted to examine the relationship between MEG3 and miR-149-3p. LncRNA MEG3 was highly expressed, whereas miR-149-3p expression was downregulated in smokers with COPD peripheral blood samples, compared with non-smokers and smokers without COPD samples. Compared with untreated human bronchial epithelial (HBE) cells, MEG3 expression was increased in cigarette smoke extract (CSE)-treated HBE cells. Compared with CSE-treated HBE cells transfected with si-NC, MEG3 knockdown promoted cell proliferation and inhibited apoptosis in CSE-treated HBE cells transfected with si-MEG3, and it also decreased the levels of IL-6, TNF-α, Bcl-2 and increased cleaved-caspase-3 and cleaved-caspase-9 in CSE-treated HBE cells transfected with si-MEG3. The luciferase assay demonstrated that miR-149-3p has target sites for MEG3. MEG3 was demonstrated to regulate the NF-κB signaling pathway by sponging miR-149-3p in CSE-treated HBE cells. In conclusion, these findings suggested that MEG3 promoted proliferation and inhibited apoptosis by regulating the NF-κB signal pathway via miR-149-3p in CSE-treated HBE cells. These results provide an insight for further verification and understanding of the molecular basis of COPD.
Collapse
|
18
|
Helal HG, Rashed MH, Abdullah OA, Salem TI, Daifalla A. MicroRNAs (−146a, −21 and −34a) are diagnostic and prognostic biomarkers for diabetic retinopathy. Biomed J 2020; 44:S242-S251. [PMID: 35304162 PMCID: PMC9068559 DOI: 10.1016/j.bj.2020.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 10/17/2020] [Accepted: 11/02/2020] [Indexed: 01/15/2023] Open
Abstract
Background Diabetic retinopathy (DR) is implicated in blindness of diabetic patients. Early diagnosis of DR is very essential to ensure good prognosis. The role of microRNAs (miRs) as biomarker diagnostic tools in DR is not fully investigated. The present study aimed to find the relation between serum relative expression of microRNAs (miR-146a, miR-21 and miR-34a) and severity of DR and to what extent their expression pattern can be used as either diagnostic or prognostic. Methods Eighty type 2 diabetic patients were classified according to severity of DR into normal, mild, moderate, severe non-proliferative diabetic retinopathy (NPDR) and proliferative diabetic retinopathy (PDR). Serum relative expressions of miRNAs were evaluated by qPCR and statistically analysed in each stage using Analysis of Variance (ANOVA) followed by Tuckey-Kramer post-test. Results Serum relative expressions of miR-146a and miR-21 were increased with increased severity of DR. miR-34a decreased with the severity of DR. The expression pattern in each group in relation to normal fundus group could be diagnostic and prognostic where miR-146a was only increased in mild group and continued with the severity. In moderate group miR-21 start to increase along with slight decrease in miR-34a. In severe NPDR group along with highly increased levels of both miR-146a and miR-21, a marked decrease in miR-34a. In PDR group miR-34a was almost diminished along with very high levels of both miR-146a and miR-21. Conclusions miRs (−146a,-21 and-34a) are promising biomarkers in DR and can help to avoid disease progression.
Collapse
Affiliation(s)
- Hend Gouda Helal
- Department of Ophthalmology, Faculty of Medicine, Benha University, Benha, Egypt.
| | - Mohammed H Rashed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Omnia Alsaied Abdullah
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Tamer Ibrahim Salem
- Department of Ophthalmology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Ahmed Daifalla
- Department of Ophthalmology, Faculty of Medicine, Benha University, Benha, Egypt
| |
Collapse
|
19
|
Makhafola TJ, Mbele M, Yacqub-Usman K, Hendren A, Haigh DB, Blackley Z, Meyer M, Mongan NP, Bates DO, Dlamini Z. Apoptosis in Cancer Cells Is Induced by Alternative Splicing of hnRNPA2/B1 Through Splicing of Bcl-x, a Mechanism that Can Be Stimulated by an Extract of the South African Medicinal Plant, Cotyledon orbiculata. Front Oncol 2020; 10:547392. [PMID: 33163396 PMCID: PMC7580256 DOI: 10.3389/fonc.2020.547392] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/21/2020] [Indexed: 01/09/2023] Open
Abstract
Alternative splicing is deregulated in cancer and alternatively spliced products can be linked to cancer hallmarks. Targeting alternative splicing could offer novel effective cancer treatments. We investigated the effects of the crude extract of a South African medicinal plant, Cotyledon orbiculata, on cell survival of colon (HCT116) and esophageal (OE33 and KYSE70) cancer cell lines. Using RNASeq, we discovered that the extract interfered with mRNA regulatory pathways. The extract caused hnRNPA2B1 to splice from the hnRNPB1 to the hnRNPA2 isoform, resulting in a switch in the BCL2L1 gene from Bcl-xL to Bcl-xS causing activation of caspase-3-cleavage and apoptosis. Similar splicing effects were induced by the known anti-cancer splicing modulator pladienolide B. Knockdown of hnRNPB1 using siRNA resulted in decreased cell viability and increased caspase-3-cleavage, and over-expression of hnRNPB1 prevented the effect of C. orbiculata extract on apoptosis and cell survival. The effect of the hnRNPA2/B1 splicing switch by the C. orbiculata extract increased hnRNPA2B1 binding to Bcl-xl/s, BCL2, MDM2, cMYC, CD44, CDK6, and cJUN mRNA. These findings suggest that apoptosis in HCT116, OE33, and KYSE cancer cells is controlled by switched splicing of hnRNPA2B1 and BCL2L1, providing evidence that hnRNPB1 regulates apoptosis. Inhibiting this splicing could have therapeutic potential for colon and esophageal cancers. Targeting hnRNPA2B1 splicing in colon cancer regulates splicing of BCL2L1 to induce apoptosis. This approach could be a useful therapeutic strategy to induce apoptosis and restrain cancer cell proliferation and tumor progression. Here, we found that the extract of Cotyledon orbiculata, a South African medicinal plant, had an anti-proliferative effect in cancer cells, mediated by apoptosis induced by alternative splicing of hnRNPA2B1 and BCL2L1.
Collapse
Affiliation(s)
- Tshepiso Jan Makhafola
- SA-Medical Research Council (MRC)/UP Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Faculty of Health Sciences, Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria, South Africa.,Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Mzwandile Mbele
- SA-Medical Research Council (MRC)/UP Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Faculty of Health Sciences, Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria, South Africa.,Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Kiren Yacqub-Usman
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Amy Hendren
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Daisy Belle Haigh
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Zoe Blackley
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Mervin Meyer
- Biolabels Unit, Department of Biotechnology, Department of Science and Technology (DST)/Mintek Nanotechnology Innovation Centre (NIC), University of the Western Cape, Bellville, South Africa
| | - Nigel Patrick Mongan
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - David Owen Bates
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Zodwa Dlamini
- SA-Medical Research Council (MRC)/UP Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Faculty of Health Sciences, Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria, South Africa
| |
Collapse
|
20
|
Nikitin PV, Ryzhova MV, Galstyan SA, Kim DS, Zubova IV, Khokhlova EA, Shugay SV. Identification of different cell clusters in the endothelium of atherosclerotic vessels and determination of inter-cluster gradient of proliferative and inflammatory activity as new diagnostic markers. Biotech Histochem 2020; 96:487-497. [PMID: 32938242 DOI: 10.1080/10520295.2020.1823016] [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: 10/23/2022] Open
Abstract
To characterize atherogenesis functionally, we studied the functional heterogeneity of endotheliocytes in carotid vessels with atherosclerotic plaques and identified several distinct cell clusters. We measured the Ki-67 labeling index (Ki-67 LI), percentage of Bcl-2 cells (CP) and expression of CCL5, IL 6 and VCAM1 in each cell cluster. We also investigated how these indicators change when the plaque becomes unstable and how they affect the risk of adverse cerebrovascular events in patients. We evaluated the inter-cluster gradient of marker activity and its relation to patient prognosis. We identified five endothelial clusters: the under plaque cluster (UPC), peripheral cluster (PC), marginal cluster (MC), transient cluster (TC) and outside plaque cluster (OC). The UPC exhibited the greatest proliferative, proinflammatory and adhesive activity, but low anti-apoptotic activity. The PC exhibited the second greatest proliferative, adhesive and proinflammatory activity. Progression of atherosclerosis and transition of a stable atherosclerotic plaque to an unstable one was accompanied by increased expression of nearly all markers. The proliferative activity in the UPC, PC and OC, and the pro-inflammatory activity in UPC and anti-apoptotic activity in the PC, were correlated with prognosis. Also, two gradients of proliferative activity and a gradient of pro-inflammatory activity were associated with risk of adverse events.
Collapse
Affiliation(s)
- P V Nikitin
- P.K. Anokhin Institute of Normal Physiology, Moscow, Russian Federation
| | - M V Ryzhova
- N. N. Burdenko National Medical Research Center of Neurosurgery, Moscow, Russian Federation
| | - S A Galstyan
- N. N. Burdenko National Medical Research Center of Neurosurgery, Moscow, Russian Federation
| | - D S Kim
- N. N. Burdenko National Medical Research Center of Neurosurgery, Moscow, Russian Federation
| | - I V Zubova
- N. N. Burdenko National Medical Research Center of Neurosurgery, Moscow, Russian Federation
| | - E A Khokhlova
- N. N. Burdenko National Medical Research Center of Neurosurgery, Moscow, Russian Federation
| | - S V Shugay
- N. N. Burdenko National Medical Research Center of Neurosurgery, Moscow, Russian Federation
| |
Collapse
|
21
|
Chong SJF, Marchi S, Petroni G, Kroemer G, Galluzzi L, Pervaiz S. Noncanonical Cell Fate Regulation by Bcl-2 Proteins. Trends Cell Biol 2020; 30:537-555. [PMID: 32307222 DOI: 10.1016/j.tcb.2020.03.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/15/2020] [Accepted: 03/19/2020] [Indexed: 12/20/2022]
Abstract
Bcl-2 proteins are widely known as key controllers of mitochondrial outer membrane permeabilization, arguably the most important step of intrinsic apoptosis. Accumulating evidence indicate that most, if not all, members of the Bcl-2 protein family also mediate a number of apoptosis-unrelated functions. Intriguingly, many of these functions ultimately impinge on cell fate decisions via apoptosis-dependent or -independent mechanisms, delineating a complex network through which Bcl-2 family members regulate cell survival and death. Here, we critically discuss the mechanisms through which Bcl-2 proteins influence cell fate as they regulate autophagy, cellular senescence, inflammation, bioenergetic metabolism, Ca2+ fluxes, and redox homeostasis.
Collapse
Affiliation(s)
| | - Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Giulia Petroni
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Guido Kroemer
- Equipe Labellisée par la Ligue Contre le Cancer, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-, HP, Paris, France; Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China; Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden; Université de Paris, Paris, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Université de Paris, Paris, France; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.
| | - Shazib Pervaiz
- Université de Paris, Paris, France; Department of Physiology, YLL School of Medicine and NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore; National University Cancer Institute, National University Health System, Singapore.
| |
Collapse
|
22
|
Kang D, Kim IR, Park YH, Im YH, Zhao D, Guallar E, Ahn JS, Cho J. Impact of a topical lotion, CG428, on permanent chemotherapy-induced alopecia in breast cancer survivors: a pilot randomized double-blind controlled clinical trial (VOLUME RCT). Support Care Cancer 2019; 28:1829-1837. [DOI: 10.1007/s00520-019-04982-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 07/04/2019] [Indexed: 12/27/2022]
|
23
|
Engineering Tumor Cells with Tumor Necrosis Factor α (TNF-α) or CD40 Ligand (CD40L) Genes Induce Anti-tumor Immune Responses. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-018-9687-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
24
|
BCL-2 family isoforms in apoptosis and cancer. Cell Death Dis 2019; 10:177. [PMID: 30792387 PMCID: PMC6384907 DOI: 10.1038/s41419-019-1407-6] [Citation(s) in RCA: 436] [Impact Index Per Article: 72.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 12/17/2018] [Accepted: 01/29/2019] [Indexed: 12/17/2022]
Abstract
The BCl-2 family has long been identified for its role in apoptosis. Following the initial discovery of BCL-2 in the context of B-cell lymphoma in the 1980s, a number of homologous proteins have since been identified. The members of the Bcl-2 family are designated as such due to their BCL-2 homology (BH) domains and involvement in apoptosis regulation. The BH domains facilitate the family members’ interactions with each other and can indicate pro- or anti-apoptotic function. Traditionally, these proteins are categorised into one of the three subfamilies; anti-apoptotic, BH3-only (pro-apoptotic), and pore-forming or ‘executioner’ (pro-apoptotic) proteins. Each of the BH3-only or anti-apoptotic proteins has a distinct pattern of activation, localisation and response to cell death or survival stimuli. All of these can vary across cell or stress types, or developmental stage, and this can cause the delineation of the roles of BCL-2 family members. Added to this complexity is the presence of relatively uncharacterised isoforms of many of the BCL-2 family members. There is a gap in our knowledge regarding the function of BCL-2 family isoforms. BH domain status is not always predictive or indicative of protein function, and several other important sequences, which can contribute to apoptotic activity have been identified. While therapeutic strategies targeting the BCL-2 family are constantly under development, it is imperative that we understand the molecules, which we are attempting to target. This review, discusses our current knowledge of anti-apoptotic BCL-2 family isoforms. With significant improvements in the potential for splicing therapies, it is important that we begin to understand the distinctions of the BCL-2 family, not limited to just the mechanisms of apoptosis control, but in their roles outside of apoptosis.
Collapse
|
25
|
Oliveira PVSD, Garcia-Rosa S, Sachetto ATA, Moretti AIS, Debbas V, De Bessa TC, Silva NT, Pereira ADC, Martins-de-Souza D, Santoro ML, Laurindo FRM. Protein disulfide isomerase plasma levels in healthy humans reveal proteomic signatures involved in contrasting endothelial phenotypes. Redox Biol 2019; 22:101142. [PMID: 30870787 PMCID: PMC6430080 DOI: 10.1016/j.redox.2019.101142] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 01/22/2019] [Accepted: 02/12/2019] [Indexed: 12/18/2022] Open
Abstract
Redox-related plasma proteins are candidate reporters of protein signatures associated with endothelial structure/function. Thiol-proteins from protein disulfide isomerase (PDI) family are unexplored in this context. Here, we investigate the occurrence and physiological significance of a circulating pool of PDI in healthy humans. We validated an assay for detecting PDI in plasma of healthy individuals. Our results indicate high inter-individual (median = 330 pg/mL) but low intra-individual variability over time and repeated measurements. Remarkably, plasma PDI levels could discriminate between distinct plasma proteome signatures, with PDI-rich (>median) plasma differentially expressing proteins related to cell differentiation, protein processing, housekeeping functions and others, while PDI-poor plasma differentially displayed proteins associated with coagulation, inflammatory responses and immunoactivation. Platelet function was similar among individuals with PDI-rich vs. PDI-poor plasma. Remarkably, such protein signatures closely correlated with endothelial function and phenotype, since cultured endothelial cells incubated with PDI-poor or PDI-rich plasma recapitulated gene expression and secretome patterns in line with their corresponding plasma signatures. Furthermore, such signatures translated into functional responses, with PDI-poor plasma promoting impairment of endothelial adhesion to fibronectin and a disturbed pattern of wound-associated migration and recovery area. Patients with cardiovascular events had lower PDI levels vs. healthy individuals. This is the first study describing PDI levels as reporters of specific plasma proteome signatures directly promoting contrasting endothelial phenotypes and functional responses.
Collapse
Affiliation(s)
- Percíllia Victória Santos de Oliveira
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Sheila Garcia-Rosa
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil; Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico, Sao Paulo, Brazil
| | | | - Ana Iochabel Soares Moretti
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Victor Debbas
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Tiphany Coralie De Bessa
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Nathalia Tenguan Silva
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Alexandre da Costa Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil; Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico, Sao Paulo, Brazil
| | | | - Francisco Rafael Martins Laurindo
- Laboratorio de Biologia Vascular, LIM-64 (Biologia Cardiovascular Translacional), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
26
|
Brett EA, Aitzetmüller MM, Sauter MA, Huemer GM, Machens HG, Duscher D. Breast cancer recurrence after reconstruction: know thine enemy. Oncotarget 2018; 9:27895-27906. [PMID: 29963246 PMCID: PMC6021250 DOI: 10.18632/oncotarget.25602] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/19/2018] [Indexed: 01/12/2023] Open
Abstract
Breast reconstruction proceeding cancer treatment carries risk, regardless of the type of surgery. From fat grafting, to flap placement, to implants, there is no guarantee that reconstruction will not stimulate breast cancer recurrence. Research in this field is clearly divided into two parts: scientific interventional studies and clinical retrospective evidence. The reconstructive procedure offers hypoxia, a wound microenvironment, bacterial load, adipose derived stem cells; agents shown experimentally to cause increased cancer cell activity. This is compelling scientific evidence which serves to bring uncertainty and fear to the reconstructive procedure. In the absence of clinical evidence, this laboratory literature landscape is now informing surgical choices. Curiously, clinical studies have not shown a clear link between breast cancer recurrence and reconstructive surgery. Where does that leave us? This review aims to analyze the science and the surgery, thereby understanding the oncological fear which accompanies breast cancer reconstruction.
Collapse
Affiliation(s)
- Elizabeth A Brett
- Department of Plastic and Hand Surgery, Technical University of Munich, Munich 81675, Germany
| | - Matthias M Aitzetmüller
- Department of Plastic and Hand Surgery, Technical University of Munich, Munich 81675, Germany
| | - Matthias A Sauter
- Department of Plastic and Hand Surgery, Technical University of Munich, Munich 81675, Germany
| | - Georg M Huemer
- Section of Plastic and Reconstructive Surgery, Kepler University Hospital Linz, Linz 4020, Austria
| | - Hans-Günther Machens
- Department of Plastic and Hand Surgery, Technical University of Munich, Munich 81675, Germany
| | - Dominik Duscher
- Department of Plastic and Hand Surgery, Technical University of Munich, Munich 81675, Germany
| |
Collapse
|
27
|
MicroRNA-326 aggravates acute lung injury in septic shock by mediating the NF-κB signaling pathway. Int J Biochem Cell Biol 2018; 101:1-11. [PMID: 29727715 DOI: 10.1016/j.biocel.2018.04.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 04/24/2018] [Accepted: 04/30/2018] [Indexed: 01/02/2023]
Abstract
Our previous studies have demonstrated that the activation of the nuclear factor-kappa B (NF-κB) signaling pathway contributes to the development of lipopolysaccharide (LPS)-induced acute lung injury (ALI) as well as an inflammatory reaction, and its inhibition may provide future therapeutic values. Thereby, this study aims to explore the effects of miR-326 on inflammatory response and ALI in mice with septic shock via the NF-κB signaling pathway. The study included normal mice and LPS-induced mouse models of septic shock with ALI. Modeled mice were transfected with the blank plasmid, miR-326 mimic, miR-326 inhibitor, si-BCL2A1 and miR-326 inhibitor + si-BCL2A1. Mean arterial pressure (MAP), airway pressure (AP), heart rate (HR) and lung wet dry (W/D) ratio were determined. Serum levels of interleukin (IL)-6, IL-10, IL-1β, and tumor necrosis factor-α (TNF-α) were detected using ELISA. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis were performed to detect the miR-326 expression and expression levels of BCL2A1, related genes of inflammatory response and the NF-κB signaling pathway in lung tissues. Cell viability and apoptosis were measured using the CCK-8 assay and flow cytometry, respectively. Compared to the ALI models and those transfected with blank plasmid, the up-regulated miR-326 expression and silenced BCL2A1 lead to decreased levels of MAP, increased AP, HR and lung W/D, increased serum levels of IL-6, IL-10, IL-1β and TNF-α, increased expressions of IL-6, IL-1β, TNF-α, NF-κB p65 (p-NF-κB p65), and iNOS with decreased expressions of BCL2A1s as well as inhibition of cell viability and enhanced cell apoptosis; the down-regulated miR-326 expression reversed the aforementioned situation. MiR-326 targeting the BCL2A1 gene activated the NF-κB signaling pathway, resulting in aggravated inflammatory response and lung injury of septic shock with ALI in mice.
Collapse
|
28
|
Silini AR, Cancelli S, Signoroni PB, Cargnoni A, Magatti M, Parolini O. The dichotomy of placenta-derived cells in cancer growth. Placenta 2017; 59:154-162. [DOI: 10.1016/j.placenta.2017.05.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/28/2017] [Accepted: 05/16/2017] [Indexed: 02/07/2023]
|
29
|
Gabellini C, Trisciuoglio D, Del Bufalo D. Non-canonical roles of Bcl-2 and Bcl-xL proteins: relevance of BH4 domain. Carcinogenesis 2017; 38:579-587. [PMID: 28203756 DOI: 10.1093/carcin/bgx016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 02/14/2017] [Indexed: 02/07/2023] Open
Abstract
Bcl-2 protein family is constituted by multidomain members originally identified as modulators of programmed cell death and whose expression is frequently misbalanced in cancer cells. The lead member Bcl-2 and its homologue Bcl-xL proteins are characterized by the presence of all four conserved BH domain and exert their antiapoptotic role mainly through the involvement of BH1, BH2 and BH3 homology domains, that mediate the interaction with the proapoptotic members of the same Bcl-2 family. The N-terminal BH4 domain of Bcl-2 and Bcl-xL is responsible for the interaction with other proteins that do not belong to Bcl-2 protein family. Beyond a classical role in inhibiting apoptosis, BH4 domain has been characterized as a crucial regulator of other important cellular functions attributed to Bcl-2 and Bcl-xL, including proliferation, autophagy, differentiation, DNA repair, cell migration, tumor progression and angiogenesis. During the last two decades a strong effort has been made to dissect the molecular pathways involved the capability of BH4 domain to regulate the canonical antiapoptotic and the non-canonical activities of Bcl-2 and Bcl-xL, creating the basis for the development of novel anticancer agents targeting this domain. Indeed, recent evidences obtained on in vitro and in vivo model of different cancer histotypes are confirming the promising therapeutic potential of BH4 domain inhibitors supporting their future employment as a novel anticancer strategy.
Collapse
Affiliation(s)
- Chiara Gabellini
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56127 Pisa, Italy
| | - Daniela Trisciuoglio
- Institute of Molecular Biology and Pathology, National Research Council, 00185 Rome, Italy and.,Preclinical Models and New Therapeutic Agents Unit, Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, Regina Elena National Cancer Institute, 00144 Rome, Italy
| |
Collapse
|
30
|
Williams EJ, Baines KJ, Berthon BS, Wood LG. Effects of an Encapsulated Fruit and Vegetable Juice Concentrate on Obesity-Induced Systemic Inflammation: A Randomised Controlled Trial. Nutrients 2017; 9:E116. [PMID: 28208713 PMCID: PMC5331547 DOI: 10.3390/nu9020116] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 01/01/2023] Open
Abstract
Phytochemicals from fruit and vegetables reduce systemic inflammation. This study examined the effects of an encapsulated fruit and vegetable (F&V) juice concentrate on systemic inflammation and other risk factors for chronic disease in overweight and obese adults. A double-blinded, parallel, randomized placebo-controlled trial was conducted in 56 adults aged ≥40 years with a body mass index (BMI) ≥28 kg/m². Before and after eight weeks daily treatment with six capsules of F&V juice concentrate or placebo, peripheral blood gene expression (microarray, quantitative polymerase chain reaction (qPCR)), plasma tumour necrosis factor (TNF)α (enzyme-linked immunosorbent assay (ELISA)), body composition (Dual-energy X-ray absorptiometry (DEXA)) and lipid profiles were assessed. Following consumption of juice concentrate, total cholesterol, low-density lipoprotein (LDL) cholesterol and plasma TNFα decreased and total lean mass increased, while there was no change in the placebo group. In subjects with high systemic inflammation at baseline (serum C-reactive protein (CRP) ≥3.0 mg/mL) who were supplemented with the F&V juice concentrate (n = 16), these effects were greater, with decreased total cholesterol, LDL cholesterol and plasma TNFα and increased total lean mass; plasma CRP was unchanged by the F&V juice concentrate following both analyses. The expression of several genes involved in lipogenesis, the nuclear factor-κB (NF-κB) and 5' adenosine monophosphate-activated protein kinase (AMPK) signalling pathways was altered, including phosphomevalonate kinase (PMVK), zinc finger AN1-type containing 5 (ZFAND5) and calcium binding protein 39 (CAB39), respectively. Therefore, F&V juice concentrate improves the metabolic profile, by reducing systemic inflammation and blood lipid profiles and, thus, may be useful in reducing the risk of obesity-induced chronic disease.
Collapse
Affiliation(s)
- Evan J Williams
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan NSW 2308, Australia.
| | - Katherine J Baines
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan NSW 2308, Australia.
| | - Bronwyn S Berthon
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan NSW 2308, Australia.
| | - Lisa G Wood
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan NSW 2308, Australia.
| |
Collapse
|
31
|
Cipollone F, Chiarelli F, Iezzi A, Fazia ML, Cuccurullo C, Pini B, De Cesare D, Torello M, Tumini S, Cuccurullo F, Mezzetti A. Relationship between Reduced BCL-2 Expression in Circulating Mononuclear Cells and Early Nephropathy in Type 1 Diabetes. Int J Immunopathol Pharmacol 2016; 18:625-35. [PMID: 16388709 DOI: 10.1177/039463200501800403] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Microalbuminuria is the earliest clinical evidence of diabetic nephropathy, but the mechanisms linking hyperglycemia and kidney complications are not clear. The aim of this study was to evaluate whether enhanced oxidative stress in patients with microalbuminuria can contribute to diabetic nephropathy development through downregulation of the antiapoptotic gene Bcl-2 that promotes in turn a pro-inflammatory status. We studied 30 patients with type 1 diabetes (15 with and 15 without microalbuminuria) compared to 15 matched healthy controls. Plasma oxidant status, and expression of Bcl-2, activated NF-kB, inducible Nitric Oxide synthase (iNOS), and monocyte chemoattractant protein (MCP)-1 in circulating monocytes were evaluated at baseline and after 8-week oral vitamin E treatment (600 mg b.i.d.). Bcl-2 expression was significantly reduced in microalbuminuric diabetic patients as a consequence of increased oxidant burden secondary to persistent hyperglycemia. Bcl-2 down-regulation was associated with enhanced expression of NF-kB, iNOS and MCP-1, and showed a strong correlation with the albumin excretion rate. Low Bcl-2 expression and high inflammatory status were normalized by vitamin E both in vivo and in vitro. Our study showed that Bcl-2 down-regulation in diabetic patients with poor glycemic control results in the activation of the NF-kB pathway leading to the development of nephropathy. Vitamin E might provide a novel form of therapy for prevention of nephropathy in diabetic patients in which an acceptable glycemic control is difficult to achieve despite insulin therapy.
Collapse
Affiliation(s)
- F Cipollone
- Atherosclerosis Prevention Center, University of Chieti G D Annunzio School of Medicine, Chieti, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Baig MS, Zaichick SV, Mao M, de Abreu AL, Bakhshi FR, Hart PC, Saqib U, Deng J, Chatterjee S, Block ML, Vogel SM, Malik AB, Consolaro MEL, Christman JW, Minshall RD, Gantner BN, Bonini MG. NOS1-derived nitric oxide promotes NF-κB transcriptional activity through inhibition of suppressor of cytokine signaling-1. ACTA ACUST UNITED AC 2015; 212:1725-38. [PMID: 26324446 PMCID: PMC4577833 DOI: 10.1084/jem.20140654] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 08/06/2015] [Indexed: 11/04/2022]
Abstract
The NF-κB pathway is central to the regulation of inflammation. Here, we demonstrate that the low-output nitric oxide (NO) synthase 1 (NOS1 or nNOS) plays a critical role in the inflammatory response by promoting the activity of NF-κB. Specifically, NOS1-derived NO production in macrophages leads to proteolysis of suppressor of cytokine signaling 1 (SOCS1), alleviating its repression of NF-κB transcriptional activity. As a result, NOS1(-/-) mice demonstrate reduced cytokine production, lung injury, and mortality when subjected to two different models of sepsis. Isolated NOS1(-/-) macrophages demonstrate similar defects in proinflammatory transcription on challenge with Gram-negative bacterial LPS. Consistently, we found that activated NOS1(-/-) macrophages contain increased SOCS1 protein and decreased levels of p65 protein compared with wild-type cells. NOS1-dependent S-nitrosation of SOCS1 impairs its binding to p65 and targets SOCS1 for proteolysis. Treatment of NOS1(-/-) cells with exogenous NO rescues both SOCS1 degradation and stabilization of p65 protein. Point mutation analysis demonstrated that both Cys147 and Cys179 on SOCS1 are required for its NO-dependent degradation. These findings demonstrate a fundamental role for NOS1-derived NO in regulating TLR4-mediated inflammatory gene transcription, as well as the intensity and duration of the resulting host immune response.
Collapse
Affiliation(s)
- Mirza Saqib Baig
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Sofia V Zaichick
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Mao Mao
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Andre L de Abreu
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Programa de Biociencias Aplicadas a Farmacia (PBF), Universidade Estadual de Maringa, Maringa 87020-900, Brazil
| | - Farnaz R Bakhshi
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Peter C Hart
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Anatomy and Cell Biology, Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN 46202
| | - Uzma Saqib
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Jing Deng
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Saurabh Chatterjee
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Michelle L Block
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Stephen M Vogel
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Asrar B Malik
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Marcia E L Consolaro
- Programa de Biociencias Aplicadas a Farmacia (PBF), Universidade Estadual de Maringa, Maringa 87020-900, Brazil
| | - John W Christman
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Richard D Minshall
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Environmental Health Sciences, University of South Carolina, Columbia, SC 29208
| | - Benjamin N Gantner
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607
| | - Marcelo G Bonini
- Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Medicine, Department of Pharmacology, Department of Anesthesiology, and Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60607 Department of Anatomy and Cell Biology, Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN 46202
| |
Collapse
|
33
|
Ciavarella S, Caselli A, Tamma AV, Savonarola A, Loverro G, Paganelli R, Tucci M, Silvestris F. A peculiar molecular profile of umbilical cord-mesenchymal stromal cells drives their inhibitory effects on multiple myeloma cell growth and tumor progression. Stem Cells Dev 2015; 24:1457-70. [PMID: 25758779 DOI: 10.1089/scd.2014.0254] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are under intensive investigation in preclinical models of cytotherapies against cancer, including multiple myeloma (MM). However, the therapeutic use of stromal progenitors holds critical safety concerns due to their potential MM-supporting activity in vivo. Here, we explored whether MSCs from sources other than BM, such as adipose tissue (AD-MSCs) and umbilical cord (UC-MSCs), affect MM cell growth in comparison to either normal (nBM-MSCs) or myelomatous marrow MSCs (MM-BM-MSCs). Results from both proliferation and clonogenic assays indicated that, in contrast to nBM- and MM-BM-MSCs, both AD and particularly UC-MSCs significantly inhibit MM cell clonogenicity and growth in vitro. Furthermore, when co-injected with UC-MSCs into mice, RPMI-8226 MM cells formed smaller subcutaneous tumor masses, while peritumoral injections of the same MSC subtype significantly delayed the tumor burden growing in subcutaneous plasmocytoma-bearing mice. Finally, both microarrays and ELISA revealed different expression of several genes and soluble factors in UC-MSCs as compared with other MSCs. Our data suggest that UC-MSCs have a distinct molecular profile that correlates with their intrinsic anti-MM activity and emphasize the UCs as ideal sources of MSCs for future cell-based therapies against MM.
Collapse
Affiliation(s)
- Sabino Ciavarella
- 1Section of Medical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Anna Caselli
- 1Section of Medical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Antonella Valentina Tamma
- 1Section of Medical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Annalisa Savonarola
- 1Section of Medical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Giuseppe Loverro
- 1Section of Medical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Roberto Paganelli
- 2Department of Medicine and Sciences of Aging, Ce.S.I. Center for Aging Studies, Stem TECH Group, University "G. D'Annunzio," Chieti Scalo, Italy
| | - Marco Tucci
- 1Section of Medical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Franco Silvestris
- 1Section of Medical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| |
Collapse
|
34
|
Simone TM, Higgins PJ. Small Molecule PAI-1 Functional Inhibitor Attenuates Vascular Smooth Muscle Cell Migration and Survival: Implications for the Therapy of Vascular Disease. NEW HORIZONS IN TRANSLATIONAL MEDICINE 2014; 2:16-19. [PMID: 25396216 PMCID: PMC4226527 DOI: 10.1016/j.nhtm.2014.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Affiliation(s)
- Tessa M Simone
- Center for Cell Biology & Cancer Research, Albany Medical College, Albany, New York 12208
| | - Paul J Higgins
- Center for Cell Biology & Cancer Research, Albany Medical College, Albany, New York 12208
| |
Collapse
|
35
|
Abstract
Atherosclerosis is the leading global cause of mortality, morbidity, and disability. Heat shock proteins (HSPs) are a highly conserved family of proteins with diverse functions expressed by all cells exposed to environmental stress. Studies have reported that several HSPs may be potential risk markers of atherosclerosis and related cardiovascular diseases, or may be directly involved in the atherogenic process itself. HSPs are expressed by cells in atherosclerotic plaque and anti-HSP has been reported to be increased in patients with vascular disease. Autoimmune responses may be generated against antigens present within the atherosclerotic plaque, including HSP and may lead to a cycle of ongoing vascular injury. It has been suggested that by inducing a state of tolerance to these antigens, the atherogenic process may be limited and thus provide a potential therapeutic approach. It has been suggested that anti-HSPs are independent predictors of risk of vascular disease. In this review, we summarize the current understanding of HSP in cardiovascular disease and highlight their potential role as diagnostic agents and therapeutic targets.
Collapse
|
36
|
Guo S, Messmer-Blust AF, Wu J, Song X, Philbrick MJ, Shie JL, Rana JS, Li J. Role of A20 in cIAP-2 protection against tumor necrosis factor α (TNF-α)-mediated apoptosis in endothelial cells. Int J Mol Sci 2014; 15:3816-33. [PMID: 24595242 PMCID: PMC3975369 DOI: 10.3390/ijms15033816] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/30/2014] [Accepted: 02/06/2014] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor α (TNF-α) influences endothelial cell viability by altering the regulatory molecules involved in induction or suppression of apoptosis. However, the underlying mechanisms are still not completely understood. In this study, we demonstrated that A20 (also known as TNFAIP3, tumor necrosis factor α-induced protein 3, and an anti-apoptotic protein) regulates the inhibitor of apoptosis protein-2 (cIAP-2) expression upon TNF-α induction in endothelial cells. Inhibition of A20 expression by its siRNA resulted in attenuating expression of TNF-α-induced cIAP-2, yet not cIAP-1 or XIAP. A20-induced cIAP-2 expression can be blocked by the inhibition of phosphatidyl inositol-3 kinase (PI3-K), but not nuclear factor (NF)-κB, while concomitantly increasing the number of endothelial apoptotic cells and caspase 3 activation. Moreover, TNF-α-mediated induction of apoptosis was enhanced by A20 inhibition, which could be rescued by cIAP-2. Taken together, these results identify A20 as a cytoprotective factor involved in cIAP-2 inhibitory pathway of TNF-α-induced apoptosis. This is consistent with the idea that endothelial cell viability is dependent on interactions between inducers and suppressors of apoptosis, susceptible to modulation by TNF-α.
Collapse
Affiliation(s)
- Shuzhen Guo
- School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Angela F Messmer-Blust
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jiaping Wu
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Xiaoxiao Song
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Melissa J Philbrick
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jue-Lon Shie
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jamal S Rana
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jian Li
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
37
|
Bodonyi-Kovacs G, Strom TB, Putheti P. A20—A Biomarker of Allograft Outcome: A Showcase in Kidney Transplantation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 809:103-16. [DOI: 10.1007/978-1-4939-0398-6_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
38
|
Stimulation of central β2-adrenoceptors suppresses NFκB activity in rat brain: A role for IκB. Neurochem Int 2013; 63:368-78. [DOI: 10.1016/j.neuint.2013.07.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 07/08/2013] [Accepted: 07/20/2013] [Indexed: 11/19/2022]
|
39
|
Guedes RP, Rocha E, Mahiou J, Moll HP, Arvelo MB, Taube JM, Peterson CR, Kaczmarek E, Longo CR, da Silva CG, Ferran C. The C-terminal domain of A1/Bfl-1 regulates its anti-inflammatory function in human endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1553-61. [PMID: 23499873 DOI: 10.1016/j.bbamcr.2013.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 02/26/2013] [Accepted: 03/01/2013] [Indexed: 11/30/2022]
Abstract
A1/Bfl-1 is a NF-κB dependent, anti-apoptotic Bcl-2 family member that contains four Bcl-2 homology domains (BH) and an amphipathic C-terminal domain, and is expressed in endothelial cells (EC). Based on NF-κB reporter assays in bovine aortic EC, we have previously demonstrated that A1, like Bcl-2 and Bcl-xL, inhibits NF-κB activation. These results, however, do not fully translate when evaluating the cell's own NF-κB machinery in human EC overexpressing A1 by means of recombinant adenovirus (rAd.) mediated gene transfer. Indeed, overexpression of full-length A1 in human umbilical vein EC (HUVEC), and human dermal microvascular EC (HDMEC) failed to inhibit NF-κB activation. However, overexpression of a mutant lacking the C-terminal domain of A1 (A1ΔC) demonstrated a potent NF-κB inhibitory effect in these cells. Disparate effects of A1 and A1ΔC on NF-κB inhibition in human EC correlated with mitochondrial (A1) versus non-mitochondrial (A1ΔC) localization. In contrast, both full-length A1 and A1ΔC protected EC from staurosporine (STS)-induced cell death, indicating that mitochondrial localization was not necessary for A1's cytoprotective function in human EC. In conclusion, our data uncover a regulatory role for the C-terminal domain of A1 in human EC: anchoring A1 to the mitochondrion, which conserves but is not necessary for its cytoprotective function, or by its absence freeing A1 from the mitochondrion and uncovering an additional anti-inflammatory effect.
Collapse
Affiliation(s)
- Renata P Guedes
- Department of Surgery, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Beclin 1 interactome controls the crosstalk between apoptosis, autophagy and inflammasome activation: impact on the aging process. Ageing Res Rev 2013; 12:520-34. [PMID: 23220384 DOI: 10.1016/j.arr.2012.11.004] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 11/28/2012] [Accepted: 11/28/2012] [Indexed: 01/10/2023]
Abstract
Autophagy and apoptosis are crucial cellular housekeeping and tissue survival mechanisms. There is emerging evidence of important crosstalk between apoptosis and autophagy which can be linked to inflammasome activation. Beclin 1 is a platform protein which assembles an interactome consisting of diverse proteins which control the initiation of autophagocytosis and distinct phases in endocytosis. Recent studies have demonstrated that the anti-apoptotic Bcl-2 family members can interact with Beclin 1 and inhibit autophagy. Consequently, impaired autophagy can trigger inflammasome activation. Interestingly, the hallmarks of the ageing process include a decline in autophagy, increased resistance to apoptosis and a low-grade inflammatory phenotype. Age-related stresses, e.g. genotoxic, metabolic and environmental insults, enhance the expression of NF-κB-driven anti-apoptotic Bcl-2 proteins which repress the Beclin 1-dependent autophagy. Suppression of autophagocytosis provokes inflammation including NF-κB activation which further potentiates anti-apoptotic defence. In a context-dependent manner, this feedback defence mechanism can enhance the aging process or provoke tumorigenesis or cellular senescence. We will review the role of Beclin 1 interactome in the crosstalk between apoptosis, autophagy and inflammasomes emphasizing that disturbances in Beclin 1-dependent autophagy can have a crucial impact on the aging process.
Collapse
|
41
|
Carbone F, Nencioni A, Mach F, Vuilleumier N, Montecucco F. Evidence on the pathogenic role of auto-antibodies in acute cardiovascular diseases. Thromb Haemost 2013; 109:854-68. [PMID: 23446994 DOI: 10.1160/th12-10-0768] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 01/29/2013] [Indexed: 02/06/2023]
Abstract
Atherothrombosis is the major determinant of acute ischaemic cardiovascular events, such as myocardial infarction and stroke. Inflammatory processes have been linked to all phases of atherogenesis In particular, the identification of autoimmunity mediators in the complex microenvironment of chronic inflammation has become the focus of attention in both early and advanced atherogenic processes. Auto-antibodies against self-molecules or new epitopes generated by oxidative processes infiltrate atherosclerotic plaques and were shown to modulate the activity of immune cells by binding various types of receptors. However, despite mounting evidence for a pathophysiological role of autoantibodies in atherothrombosis, the clinical relevance for circulating autoantibodies in cardiovascular outcomes is still debated. This review aims at illustrating the mechanisms by which different types of autoantibodies might either promote or repress atherothrombosis and to discuss the clinical studies assessing the role of auto-antibodies as prognostic biomarkers of plaque vulnerability.
Collapse
Affiliation(s)
- F Carbone
- Cardiology Division, Department of Medicine, Geneva University Hospital, Foundation for Medical Researches, 64 Avenue Roseraie, Geneva, Switzerland
| | | | | | | | | |
Collapse
|
42
|
Heat shock proteins: pathogenic role in atherosclerosis and potential therapeutic implications. Autoimmune Dis 2012; 2012:502813. [PMID: 23304456 PMCID: PMC3530228 DOI: 10.1155/2012/502813] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 09/15/2012] [Accepted: 09/24/2012] [Indexed: 11/17/2022] Open
Abstract
Heat shock proteins (HSPs) are a highly conserved group of proteins that are constitutively expressed and function as molecular chaperones, aiding in protein folding and preventing the accumulation of misfolded proteins. In the arterial wall, HSPs have a protective role under normal physiologic conditions. In disease states, however, HSPs expressed on the vascular endothelial cell surface can act as targets for detrimental autoimmunity due to their highly conserved sequences. Developing therapeutic strategies for atherosclerosis based on HSPs is challenged by the need to balance such physiologic and pathologic roles of these proteins. This paper summarizes the role of HSPs in normal vascular wall processes as well as in the development and progression of atherosclerosis. The potential implications of HSPs in clinical therapies for atherosclerosis are also discussed.
Collapse
|
43
|
Charreau B. Signaling of endothelial cytoprotection in transplantation. Hum Immunol 2012; 73:1245-52. [DOI: 10.1016/j.humimm.2012.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 06/25/2012] [Accepted: 07/09/2012] [Indexed: 12/22/2022]
|
44
|
Xu Q, Metzler B, Jahangiri M, Mandal K. Molecular chaperones and heat shock proteins in atherosclerosis. Am J Physiol Heart Circ Physiol 2011; 302:H506-14. [PMID: 22058161 DOI: 10.1152/ajpheart.00646.2011] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In response to stress stimuli, mammalian cells activate an ancient signaling pathway leading to the transient expression of heat shock proteins (HSPs). HSPs are a family of proteins serving as molecular chaperones that prevent the formation of nonspecific protein aggregates and assist proteins in the acquisition of their native structures. Physiologically, HSPs play a protective role in the homeostasis of the vessel wall but have an impact on immunoinflammatory processes in pathological conditions involved in the development of atherosclerosis. For instance, some members of HSPs have been shown to have immunoregulatory properties and modification of innate and adaptive response to HSPs, and can protect the vessel wall from the disease. On the other hand, a high degree of sequence homology between microbial and mammalian HSPs, due to evolutionary conservation, carries a risk of misdirected autoimmunity against HSPs expressed on the stressed cells of vascular endothelium. Furthermore, HSPs and anti-HSP antibodies have been shown to elicit production of proinflammatory cytokines. Potential therapeutic use of HSP in prevention of atherosclerosis involves achieving optimal balance between protective and immunogenic effects of HSPs and in the progress of research on vaccination. In this review, we update the progress of studies on HSPs and the integrity of the vessel wall, discuss the mechanism by which HSPs exert their role in the disease development, and highlight the potential clinic translation in the research field.
Collapse
Affiliation(s)
- Qingbo Xu
- Cardiovascular Division, King's British Heart Foundation Center, King's College London, London, United Kingdom
| | | | | | | |
Collapse
|
45
|
Das S, Babick AP, Xu YJ, Takeda N, Rodriguez-Levya D, Dhalla NS. TNF-alpha-mediated signal transduction pathway is a major determinant of apoptosis in dilated cardiomyopathy. J Cell Mol Med 2010; 14:1988-97. [PMID: 19754666 PMCID: PMC3823280 DOI: 10.1111/j.1582-4934.2009.00904.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 08/27/2009] [Indexed: 12/05/2022] Open
Abstract
Although J2N-k strain of cardiomyopathic hamsters is an excellent model of dilated cardiomyopathy, the presence and mechanisms of apoptosis in the hearts of these genetically modified animals have not been investigated. This study examined the hypothesis that cardiac dysfunction and apoptosis in the cardiomyopathic hamsters were associated with tumour necrosis factor-alpha (TNF-alpha)-mediated signalling pathway involving the activation of some pro-apoptotic proteins and/or deactivation of some antiapoptotic proteins. Echocardiographic assessment of 31-week-old hamsters indicated an increase in the internal dimension of the left ventricle as well as decreases in the ejection fraction, fractional shortening and cardiac output without any evidence of cardiac hypertrophy. Increased level of TNF-alpha and apoptosis in cardiomyopathic hearts were accompanied by increased protein content for protein kinase C (PKC) -alpha and -epsilon isozymes as well as caspases 3 and 9. Phosphorylated protein content for p38 MAPK and NF kappaB was increased whereas that for Erk1/2, BAD and Bcl-2 was decreased in cardiomyopathic hearts. These results support the view that TNF-alpha and PKC isozymes may promote apoptosis due to the activation of p38 MAPK and deactivation of Erk1/2 pathways, and these changes may contribute toward the development of cardiac dysfunction in dilated cardiomyopathy.
Collapse
Affiliation(s)
- Samarjit Das
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Center and Department of Physiology, Faculty of Medicine, University of ManitobaWinnipeg, Canada
| | - Andrea P Babick
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Center and Department of Physiology, Faculty of Medicine, University of ManitobaWinnipeg, Canada
| | - Yan-Jun Xu
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Center and Department of Physiology, Faculty of Medicine, University of ManitobaWinnipeg, Canada
| | - Nobuakira Takeda
- Department of General Medicine, Aoto Hospital, Jikei University School of MedicineTokyo, Japan
| | - Delfin Rodriguez-Levya
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Center and Department of Physiology, Faculty of Medicine, University of ManitobaWinnipeg, Canada
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Center and Department of Physiology, Faculty of Medicine, University of ManitobaWinnipeg, Canada
| |
Collapse
|
46
|
Kern TS, Du Y, Miller CM, Hatala DA, Levin LA. Overexpression of Bcl-2 in vascular endothelium inhibits the microvascular lesions of diabetic retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2550-8. [PMID: 20363911 DOI: 10.2353/ajpath.2010.091062] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recent studies on the pathogenesis of diabetic retinopathy have focused on correcting adverse biochemical alterations, but there have been fewer efforts to enhance prosurvival pathways. Bcl-2 is the archetypal member of a group of antiapoptotic proteins. In this study, we investigated the ability of overexpressing Bcl-2 in vascular endothelium to protect against early stages of diabetic retinopathy. Transgenic mice overexpressing Bcl-2 regulated by the pre-proendothelin promoter were generated, resulting in increased endothelial Bcl-2. Diabetes was induced with streptozotocin, and mice were sacrificed at 2 months of study to measure superoxide generation, leukostasis, and immunohistochemistry, and at 7 months to assess retinal histopathology. Diabetes of 2 months duration caused a significant decrease in expression of Bcl-2 in retina, upregulation of Bax in whole retina and isolated retinal microvessels, and increased generation of retinal superoxide and leukostasis. Seven months of diabetes caused a significant increase in the number of degenerate (acellular) capillaries in diabetic animals. Furthermore, overexpression of Bcl-2 in the vascular endothelium inhibited the diabetes-induced degeneration of retinal capillaries and aberrant superoxide generation, but had no effect on Bax expression or leukostasis. Therefore, overexpression of Bcl-2 in endothelial cells inhibits the capillary degeneration that is characteristic of the early stages of diabetic retinopathy, and this effect seems likely to involve inhibition of oxidative stress.
Collapse
Affiliation(s)
- Timothy S Kern
- Center for Diabetes Research, Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | | | | | | | | |
Collapse
|
47
|
Uchida Y, Ke B, Freitas MCS, Ji H, Zhao D, Benjamin ER, Najafian N, Yagita H, Akiba H, Busuttil RW, Kupiec-Weglinski JW. The emerging role of T cell immunoglobulin mucin-1 in the mechanism of liver ischemia and reperfusion injury in the mouse. Hepatology 2010; 51:1363-72. [PMID: 20091883 PMCID: PMC3066468 DOI: 10.1002/hep.23442] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The T cell immunoglobulin and mucin domain-containing molecules (TIM) protein family, which is expressed by T cells, plays a crucial role in regulating host adaptive immunity and tolerance. However, its role in local inflammation, such as innate immunity-dominated organ ischemia-reperfusion injury (IRI), remains unknown. Liver IRI occurs frequently after major hepatic resection or liver transplantation. Using an antagonistic anti-TIM-1 antibody (Ab), we studied the role of TIM-1 signaling in the model of partial warm liver ischemia followed by reperfusion. Anti-TIM-1 Ab monotherapy ameliorated the hepatocellular damage and improved liver function due to IR, as compared with controls. Histological examination has revealed that anti-TIM-1 Ab treatment decreased local neutrophil infiltration, inhibited sequestration of T lymphocytes, macrophages, TIM-1 ligand-expressing TIM-4(+) cells, and reduced liver cell apoptosis. Intrahepatic neutrophil activity and induction of proinflammatory cytokines/chemokines were also reduced in the treatment group. In parallel in vitro studies, anti-TIM-1 Ab suppressed interferon-gamma (IFN-gamma) production in concanavalin A (conA)-stimulated spleen T cells, and diminished tumor necrosis factor alpha (TNF-alpha)/interleukin (IL)-6 expression in a macrophage/spleen T cell coculture system. This is the first study to provide evidence for the novel role of TIM-1 signaling in the mechanism of liver IRI. TIM-1 regulates not only T for the role of cell activation but may also affect macrophage function in the local inflammation response. These results provide compelling data for further investigation of TIM-1 pathway in the mechanism of IRI, to improve liver function, expand the organ donor pool, and improve the overall success of liver transplantation.
Collapse
Affiliation(s)
- Yoichiro Uchida
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Bibo Ke
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Maria Cecilia S Freitas
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Haofeng Ji
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Danyun Zhao
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Elizabeth R Benjamin
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Nader Najafian
- Transplantation Research Center, Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Hisaya Akiba
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ronald W Busuttil
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Jerzy W. Kupiec-Weglinski
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA,Address correspondence to: Jerzy W. Kupiec-Weglinski, MD, PhD. Dumont - UCLA Transplant Center 77-120 CHS, 10833 Le Conte Ave, Los Angeles, CA 90095. Phone: (310) 825-4196; Fax: (310) 267-2358;
| |
Collapse
|
48
|
Zhao J, Bolton EM, Bradley JA, Lever AML. Lentiviral-mediated overexpression of Bcl-xL protects primary endothelial cells from ischemia/reperfusion injury-induced apoptosis. J Heart Lung Transplant 2010; 28:936-43. [PMID: 19716047 DOI: 10.1016/j.healun.2009.05.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Revised: 04/14/2009] [Accepted: 05/08/2009] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Endothelial cells (EC) respond to mild injurious stimuli by upregulating anti-apoptotic gene expression to maintain endothelial integrity. EC dysfunction and apoptosis resulting from ischemia/reperfusion injury may contribute to chronic allograft rejection. We optimized conditions for lentiviral vector (LVV) transduction of rat aortic endothelial cells (RAEC) and investigated whether LVV delivery of the anti-apoptotic gene, Bcl-xL, protects RAEC from apoptotic death using in vitro models of hypoxia and ischemia/reperfusion injury. METHODS LVV containing Bcl-xL were generated from a human immunodeficiency virus (HIV)-1 construct. EC were prepared from rat aorta. Hypoxia/reperfusion (H/R) or ischemia/reperfusion (I/R) injury was induced in vitro and apoptosis was assessed using caspase-3 activity, Annexin V/PI and TUNEL staining. RESULTS After in vitro induction of H/R or I/R injury, RAEC showed duration-dependent apoptosis. We confirmed the damaging effect of the reperfusion phase. Endogenous Bax expression increased with I/R injury, whereas endogenous Bcl-xL remained constant. RAEC transduced with LVV expressing Bcl-xL were protected from early apoptosis caused by I/R injury, correlating with reduced cytochrome c release into the cytosol. CONCLUSIONS Overexpressing Bcl-xL protects RAEC from I/R injury. This protective effect may be attributed to altering the balance of pro- and anti-apoptotic proteins, resulting in sequestration of the harmful Bax protein, and may open up new strategies for controlling chronic allograft rejection.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | | | | | | |
Collapse
|
49
|
Laurence JM, Allen RDM, McCaughan GW, Logan GJ, Alexander IE, Bishop GA, Sharland AF. Gene therapy in transplantation. Transplant Rev (Orlando) 2009; 23:159-70. [PMID: 19428235 DOI: 10.1016/j.trre.2009.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gene therapy is an exciting and novel technology that offers the prospect of improving transplant outcomes beyond those achievable with current clinical protocols. This review explores both the candidate genes and ways in which they have been deployed to overcome both immune and non-immune barriers to transplantation success in experimental models. Finally, the major obstacles to implementing gene therapy in the clinic are considered.
Collapse
Affiliation(s)
- Jerome M Laurence
- Collaborative Transplantation Research Group, Bosch Insitute, Royal Prince Alfred Hospital and University of Sydney, NSW 2006, Australia
| | | | | | | | | | | | | |
Collapse
|
50
|
Biologic sequelae of I{kappa}B kinase (IKK) inhibition in multiple myeloma: therapeutic implications. Blood 2009; 113:5228-36. [PMID: 19270264 DOI: 10.1182/blood-2008-06-161505] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Nuclear factor-kappaB (NF-kappaB) has an important role in multiple myeloma (MM) cell pathogenesis in the context of the bone marrow (BM) microenvironment. In NF-kappaB signaling cascades, IkappaB kinase alpha (IKKalpha) and IKKbeta are key molecules that predominantly mediate noncanonical and canonical pathways, respectively. In this study, we examined the biologic sequelae of the inhibition of IKKalpha versus IKKbeta in MM cell lines. All MM cell lines have constitutive canonical NF-kappaB activity, and a subset of MM cell lines shows noncanonical NF-kappaB activity. Adhesion to BM stromal cells further activates both canonical and noncanonical NF-kappaB activity. IKKbeta inhibitor MLN120B blocks canonical pathway and growth of MM cell lines but does not inhibit the noncanonical NF-kappaB pathway. Although IKKalpha knockdown induces significant growth inhibition in the cell lines with both canonical and noncanonical pathways, it does not inhibit NF-kappaB activation. Importantly, IKKalpha down-regulation decreases expression of beta-catenin and aurora-A, which are known to mediate MM cell growth and survival. Finally, IKKbeta inhibitor enhances the growth inhibition triggered by IKKalpha down-regulation in MM cells with both canonical and noncanonical NF-kappaB activity. Combination therapy targeting these kinases therefore represents a promising treatment strategy in MM.
Collapse
|