1
|
Giagulli VA, Lisco G, De Tullio A, Guastamacchia E, Triggiani V, Jirillo E. The pathogenic role of the immune system in erectile dysfunction and Peyronie's disease: focusing on immunopathophysiology and potential therapeutic strategies. Sex Med Rev 2024; 12:210-220. [PMID: 38196188 DOI: 10.1093/sxmrev/qead055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/25/2023] [Accepted: 10/09/2023] [Indexed: 01/11/2024]
Abstract
INTRODUCTION Erectile dysfunction (ED) represents the major cause of male sexual dysfunction, which is often associated with obesity, diabetes mellitus, atherosclerotic cardiovascular disease, and cigarette smoking. Peyronie's disease is a chronic disorder associated with irreversible fibrotic damage of the tunica albuginea leading to ED, painful erection, coital disturbance, and physical and social complaints. Both conditions are characterized by chronic inflammation, oxidative stress, and significant changes in intracavernous hydrodynamics. In this scenario, oxidized lipoproteins, M1-polarized macrophages, proinflammatory cytokines (such as the tumor necrosis factor α), endothelial nitric oxide synthase, penile smooth muscle cells, and toll-like receptors represent the main triggers of the inflammatory process in ED. Phosphodiesterase-5 inhibitors are the most common treatment for ED. This treatment is used intermittently, as it is conceived as a symptomatic and not curative therapy. Moreover, not all patients respond to phosphodiesterase-5 inhibitors (35%-85%), particularly those with dysmetabolic phenotypes. Additional or alternative treatments are therefore desirable, mostly in refractory cases. OBJECTIVES In this review, we describe the immune-mediated pathogenesis of ED and Peyronie's disease (PD). In our literature search we placed particular emphasis on potentially practical therapeutic approaches, including natural products (such as polyphenols), due to their anti-inflammatory and antioxidant activities, stem cell therapy, and platelet-derived preparations. METHODS We searched PubMed/MEDLINE, Web of Science, Scopus, Cochrane Library, Google Scholar, and institutional websites. Original studies, narrative reviews, systematic reviews, and meta-analyses written in English were searched, screened, and selected. RESULTS In animal models of ED and PD, therapeutic approaches, including anti-inflammatory and antioxidant agents, stem cell therapy, and platelet-derived preparations, have provided positive results, including improved penile function, reduced inflammation and oxidative stress, and promotion of tissue repair. However, clinical evidence of improvement in human patients is still insufficient. CONCLUSION Promising results for treating ED and PD have been shown in preclinical and pilot clinical studies, but specific clinical trials are needed to validate the efficacy of these therapeutic approaches in men with ED.
Collapse
Affiliation(s)
- Vito Angelo Giagulli
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology, and Rare Diseases, School of Medicine, University of Bari "Aldo Moro", Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Giuseppe Lisco
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology, and Rare Diseases, School of Medicine, University of Bari "Aldo Moro", Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Anna De Tullio
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology, and Rare Diseases, School of Medicine, University of Bari "Aldo Moro", Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Edoardo Guastamacchia
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology, and Rare Diseases, School of Medicine, University of Bari "Aldo Moro", Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology, and Rare Diseases, School of Medicine, University of Bari "Aldo Moro", Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Emilio Jirillo
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology, and Rare Diseases, School of Medicine, University of Bari "Aldo Moro", Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Italy
| |
Collapse
|
2
|
Zhan T, Chen Y, Dong L, Wei T, Lu D, Wang Q, Wu Z, Xu J, Ge M. Nicotinamide phosphoribose transferase facilitates macrophage-mediated pulmonary fibrosis through the Sirt1-Smad7 pathway in mice. Eur J Pharmacol 2024; 967:176355. [PMID: 38280463 DOI: 10.1016/j.ejphar.2024.176355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/27/2023] [Accepted: 01/23/2024] [Indexed: 01/29/2024]
Abstract
Pulmonary fibrosis is a challenging lung disease characterized by a bleak prognosis. A pivotal element in the progression of this disease is the dysregulated recruitment of macrophages. Nicotinamide phosphoribose transferase (NAMPT), secreted by alveolar epithelial cells and inflammatory cells, has been previously identified to influence macrophage inflammation in acute lung injury through the nicotinamide adenine dinucleotide (NAD) rescue synthesis pathway. Nonetheless, the exact role of NAMPT in the regulation of lung fibrosis is yet to be elucidated. In our research, we employed bleomycin (BLM) to induce pulmonary fibrosis in Namptflox/flox;Cx3cr1CreER mice, using Namptflox/flox mice as controls. Our findings revealed an augmented expression of NAMPT concurrent with a marked increase in the secretion of NAD and inflammatory cytokines such as IL-6, TNF-α, and TGF-β1 post-BLM treatment. Furthermore, an upsurge in NAMPT-positive macrophages was observed in the lungs of BLM-treated Namptflox/flox mice. Notably, a conditional knockout of NAMPT (NAMPT cKO) in lung macrophages curtailed the BLM-induced inflammatory responses and significantly mitigated pulmonary fibrosis. This was associated with diminished phospho-Sirt1 (p-Sirt1) expression levels and a concomitant rise in mothers against decapentaplegic homolog 7 (Smad7) expression in BLM-treated mouse lungs and murine RAW 264.7 macrophage cells. Collectively, our data suggests that NAMPT exacerbates macrophage-driven inflammation and pulmonary fibrosis via the Sirt1-Smad7 pathway, positioning NAMPT as a promising therapeutic target for pulmonary fibrosis intervention.
Collapse
Affiliation(s)
- Tianwei Zhan
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), 158 Shang-Tang Road, 310014, Hangzhou, Zhejiang, People's Republic of China
| | - Yaling Chen
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yu-Hang-Tang Road, 310058, Hangzhou, Zhejiang, People's Republic of China
| | - Lingjun Dong
- Department of Thoracic Surgery, Shaoxing People's Hospital, 568-Zhongxing North Road, 312000, Shaoxing, Zhejiang, People's Republic of China
| | - Taofeng Wei
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yu-Hang-Tang Road, 310058, Hangzhou, Zhejiang, People's Republic of China
| | - Dongning Lu
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), 158 Shang-Tang Road, 310014, Hangzhou, Zhejiang, People's Republic of China
| | - Qi Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jie-Fang Road, 310009, Hangzhou, Zhejiang, People's Republic of China
| | - Zixiang Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jie-Fang Road, 310009, Hangzhou, Zhejiang, People's Republic of China
| | - Jiajie Xu
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), 158 Shang-Tang Road, 310014, Hangzhou, Zhejiang, People's Republic of China.
| | - Minghua Ge
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), 158 Shang-Tang Road, 310014, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
3
|
van der Geest R, Peñaloza HF, Xiong Z, Gonzalez-Ferrer S, An X, Li H, Fan H, Tabary M, Nouraie SM, Zhao Y, Zhang Y, Chen K, Alder JK, Bain WG, Lee JS. BATF2 enhances proinflammatory cytokine responses in macrophages and improves early host defense against pulmonary Klebsiella pneumoniae infection. Am J Physiol Lung Cell Mol Physiol 2023; 325:L604-L616. [PMID: 37724373 PMCID: PMC11068429 DOI: 10.1152/ajplung.00441.2022] [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: 12/29/2022] [Revised: 07/12/2023] [Accepted: 08/31/2023] [Indexed: 09/20/2023] Open
Abstract
Basic leucine zipper transcription factor ATF-like 2 (BATF2) is a transcription factor that is emerging as an important regulator of the innate immune system. BATF2 is among the top upregulated genes in human alveolar macrophages treated with LPS, but the signaling pathways that induce BATF2 expression in response to Gram-negative stimuli are incompletely understood. In addition, the role of BATF2 in the host response to pulmonary infection with a Gram-negative pathogen like Klebsiella pneumoniae (Kp) is not known. We show that induction of Batf2 gene expression in macrophages in response to Kp in vitro requires TRIF and type I interferon (IFN) signaling, but not MyD88 signaling. Analysis of the impact of BATF2 deficiency on macrophage effector functions in vitro showed that BATF2 does not directly impact macrophage phagocytic uptake and intracellular killing of Kp. However, BATF2 markedly enhanced macrophage proinflammatory gene expression and Kp-induced cytokine responses. In vivo, Batf2 gene expression was elevated in lung tissue of wild-type (WT) mice 24 h after pulmonary Kp infection, and Kp-infected BATF2-deficient (Batf2-/-) mice displayed an increase in bacterial burden in the lung, spleen, and liver compared with WT mice. WT and Batf2-/- mice showed similar recruitment of leukocytes following infection, but in line with in vitro observations, proinflammatory cytokine levels in the alveolar space were reduced in Batf2-/- mice. Altogether, these results suggest that BATF2 enhances proinflammatory cytokine responses in macrophages in response to Kp and contributes to the early host defense against pulmonary Kp infection.NEW & NOTEWORTHY This study investigates the signaling pathways that mediate induction of BATF2 expression downstream of TLR4 and also the impact of BATF2 on the host defense against pulmonary Kp infection. We demonstrate that Kp-induced upregulation of BATF2 in macrophages requires TRIF and type I IFN signaling. We also show that BATF2 enhances Kp-induced macrophage cytokine responses and that BATF2 contributes to the early host defense against pulmonary Kp infection.
Collapse
Affiliation(s)
- Rick van der Geest
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Hernán F Peñaloza
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Zeyu Xiong
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Shekina Gonzalez-Ferrer
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Xiaojing An
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Huihua Li
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Hongye Fan
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Mohammadreza Tabary
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - S Mehdi Nouraie
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Yanwu Zhao
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Yingze Zhang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Kong Chen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Jonathan K Alder
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - William G Bain
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, United States
| | - Janet S Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Acute Lung Injury Center of Excellence, Department of Medicine, Pittsburgh, Pennsylvania, United States
- Division of Pulmonary and Critical Care Medicine, John T. Milliken Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| |
Collapse
|
4
|
Hwang J, Jang S, Kim C, Lee S, Jeong HS. Role of Stem Cell-Derived Exosomes and microRNAs in Spinal Cord Injury. Int J Mol Sci 2023; 24:13849. [PMID: 37762150 PMCID: PMC10530823 DOI: 10.3390/ijms241813849] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Neurological disorders represent a global health problem. Current pharmacological treatments often lead to short-term symptomatic relief but have dose-dependent side effects, such as inducing orthostatic arterial hypotension due to the blockade of alpha receptors, cardiotoxic effects due to impaired repolarization, and atrioventricular block and tachycardia, including ventricular fibrillation. These challenges have driven the medical community to seek effective treatments for this serious global health threat. Mesenchymal stem cells (MSCs) are pluripotent cells with anti-inflammatory, anti-apoptotic, and immunomodulatory properties, providing a promising alternative due to their ability to differentiate, favorable culture conditions, in vitro manipulation ability, and robust properties. Although MSCs themselves rarely differentiate into neurons at the site of injury after transplantation in vivo, paracrine factors secreted by MSCs can create environmental conditions for cell-to-cell communication and have shown therapeutic effects. Recent studies have shown that the pleiotropic effects of MSCs, particularly their immunomodulatory potential, can be attributed primarily to these paracrine factors. Exosomes derived from MSCs are known to play an important role in these effects. Many studies have evaluated the potential of exosome-based therapies for the treatment of various neurological diseases. In addition to exosomes, various miRNAs derived from MSCs have been identified to regulate genes and alleviate neuropathological changes in neurodegenerative diseases. This review explores the burgeoning field of exosome-based therapies, focusing on the effects of MSC-derived exosomes and exosomal miRNAs, and summarizes recent findings that shed light on the potential of exosomes in the treatment of neurological disorders. The insights gained from this review may pave the way for innovative and effective treatments for these complex conditions. Furthermore, we suggest the therapeutic effects of exosomes and exosomal miRNAs from MSCs, which have a rescue potential in spinal cord injury via diverse signaling pathways.
Collapse
Affiliation(s)
- Jinsu Hwang
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; (J.H.); (S.J.)
| | - Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; (J.H.); (S.J.)
| | - Choonghyo Kim
- Department of Neurosurgery, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea;
| | - Sungjoon Lee
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea;
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; (J.H.); (S.J.)
| |
Collapse
|
5
|
Žaloudíková M. Mechanisms and Effects of Macrophage Polarization and Its Specifics in Pulmonary Environment. Physiol Res 2023; 72:S137-S156. [PMID: 37565418 PMCID: PMC10660583 DOI: 10.33549/physiolres.935058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 06/09/2023] [Indexed: 12/01/2023] Open
Abstract
Macrophages are a specific group of cells found in all body tissues. They have specific characteristics in each of the tissues that correspond to the functional needs of the specific environment. These cells are involved in a wide range of processes, both pro-inflammatory and anti-inflammatory ("wound healing"). This is due to their specific capacity for so-called polarization, a phenotypic change that is, moreover, partially reversible compared to other differentiated cells of the human body. This promises a wide range of possibilities for its influence and thus therapeutic use. In this article, we therefore review the mechanisms that cause polarization, the basic classification of polarized macrophages, their characteristic markers and the effects that accompany these phenotypic changes. Since the study of pulmonary (and among them mainly alveolar) macrophages is currently the focus of scientific interest of many researchers and these macrophages are found in very specific environments, given mainly by the extremely high partial pressure of oxygen compared to other locations, which specifically affects their behavior, we will focus our review on this group.
Collapse
Affiliation(s)
- M Žaloudíková
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
6
|
Mueen RM, Al-Juaifari M, Abosaooda M, Qassam H, Hadi NR. Lung protective effect of Ticagrelor in endotoxemia. J Med Life 2023; 16:941-947. [PMID: 37675176 PMCID: PMC10478651 DOI: 10.25122/jml-2022-0308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/03/2023] [Indexed: 09/08/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. This study aimed to investigate the potential protective effect of the lungs in sepsis by modulating inflammatory and oxidative stress markers. Twenty-four adult male Swiss-albino mice, aged 8-12 weeks and weighing 20-30 g, were divided into four equal groups (n=6): sham (laparotomy only), CLP (laparotomy plus cecal ligation and puncture), vehicle (DMSO administered one hour before CLP), and Ticagrelor (50 mg/kg IP administered one hour before CLP). Tissue levels of pro-inflammatory and oxidative stress markers in the lung were assessed using ELISA. F2 isoprostane levels were significantly higher in the sepsis group (p<0.05) compared to the sham group, while Ticagrelor significantly decreased the inflammatory and oxidative stress markers compared to the sepsis group. All mice in the sepsis group had considerable (p=0.05) lung tissue damage, but Ticagrelor considerably decreased lung tissue injury (p=0.05). Furthermore, Ticagrelor was found to reduce tissue cytokine levels of the lung (IL-1, TNF a, IL-6, F2 isoprostane, GPR 17, MIF) in male mice during CLP-induced polymicrobial sepsis by modulation of pro-inflammatory and oxidative stress cascade signaling pathways.
Collapse
Affiliation(s)
- Ruaa Murtada Mueen
- Department of Pharmacology & Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | - Maytham Al-Juaifari
- KMG Klinikum Güstrow, Clinic for Trauma Surgery, Spinal Surgery and Orthopedics, Güstrow, Germany
| | | | - Heider Qassam
- Department of Pharmacology, Faculty of Medicine, University of Kufa, Iraq
| | - Najah Rayish Hadi
- Department of Pharmacology & Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| |
Collapse
|
7
|
Castiglione F, Albersen M, Fiorenzo S, Hedlund P, Cakir OO, Pavone C, Alnajjar HM, Joniau S, Muneer A. Long-term consequences of bilateral cavernous crush injury in normal and diabetic rats: a functional study. Int J Impot Res 2022; 34:781-785. [PMID: 34611324 DOI: 10.1038/s41443-021-00474-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/28/2021] [Accepted: 09/20/2021] [Indexed: 01/06/2023]
Abstract
A recent statement from the European-Society-for-Sexual-Medicine has highlighted the limitations of using the rat model for nerve-sparing prostatectomy. The use of young rats with no comorbidities and the early evaluation of the erectile function (EF) are deemed a source of bias. Our aim was to evaluate the long-term consequences in EF of bilateral nerve cavernous crush- injury (BNCI) in type 1 diabetic (DM) rats 30-male/12-week-old rats were divided into four groups: Sham, BNCI, DM, and BNCI + DM. Sham group underwent an intraperitoneal injection (IP) of saline solution and after 1 month underwent a sham laparotomy. BNCI underwent an IP of saline solution and after 1 month to BNCI. DM underwent an IP of 60 mg/kg-1-streptozotocin (STZ) and after 1 month to a sham laparotomy. BNCI + DM underwent an IP of 60 mg/kg-1-STZ and after 1 month to BNCI. After 5 months from the induction of diabetes, all rats underwent measurement of intracorporeal pressure (ICP) and mean arterial pressure (MAP) during CN-electrostimulation. Multiple groups were compared using Kruskal-Wallis one-way analysis of variance followed by Mann-Whitney U test for post hoc comparisons. Blood glucose-level was higher (p < 0.05) in the groups with DM and BNCI + DM. After 5-months, DM and BNCI + DM also showed a lower weight compared to other groups (p < 0.05). No differences were noted in ICP/MAP between the sham and BNCI. BNCI + DM showed lower ICP/MAP compared to all the groups (p < 0.05). DM Showed lower ICP/MAP compared to Sham and BNCI (p < 0.05). BNCI in rats without comorbidities did not induce long-term erectile dysfunction (ED) suggesting a spontaneous EF recovery. BNCI in DM induced long-term ED. The results of previous short-term studies can only provide evidence on the time to recovery of spontaneous EF as to the actual EF recovery rate.
Collapse
Affiliation(s)
- Fabio Castiglione
- Department of Urology, University College London Hospital, London, UK. .,Division of Surgery and Interventional Science, University College London, London, UK. .,Division of Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - Maarten Albersen
- Laboratory for Experimental Urology, Organ systems, Department of Development and Regeneration, University of Leuven, Leuven, Belgium
| | - Salvatore Fiorenzo
- Section of Urology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Petter Hedlund
- Department of Clinical and Experimental Pharmacology, Lund University, Lund, Sweden.,Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Omer Onur Cakir
- Department of Urology, University College London Hospital, London, UK
| | - Carlo Pavone
- Section of Urology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | | | - Steven Joniau
- Laboratory for Experimental Urology, Organ systems, Department of Development and Regeneration, University of Leuven, Leuven, Belgium
| | - Asif Muneer
- Department of Urology, University College London Hospital, London, UK.,Division of Surgery and Interventional Science, University College London, London, UK.,Division of Surgery and Interventional Science and NIHR Biomedical Research Centre University College London Hospital, London, UK
| | | |
Collapse
|
8
|
Sun W, Wang X, Wang D, Lu L, Lin H, Zhang Z, Jia Y, Nie X, Liu T, Fu W. CD40×HER2 bispecific antibody overcomes the CCL2-induced trastuzumab resistance in HER2-positive gastric cancer. J Immunother Cancer 2022; 10:jitc-2022-005063. [PMID: 35851310 PMCID: PMC9295658 DOI: 10.1136/jitc-2022-005063] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2022] [Indexed: 02/06/2023] Open
Abstract
Background There was much hard work to study the trastuzumab resistance in HER2-positive gastric cancer (GC), but the information which would reveal this abstruse mechanism is little. In this study, we aimed to investigate the roles of tumor cell-derived CCL2 on trastuzumab resistance and overcome the resistance by treatment with the anti-CD40-scFv-linked anti-HER2 (CD40 ×HER2) bispecific antibody (bsAb). Methods We measured the levels of CCL2 expression in HER2-positive GC tissues, and revealed biological functions of tumor cell-derived CCL2 on tumor-associated macrophages (TAMs) and the trastuzumab resistance. Then, we developed CD40 ×HER2 bsAb, and examined the targeting roles on HER2 and CD40, to overcome the trastuzumab resistance without systemic toxicity. Results We found the level of CCL2 expression in HER2-postive GC was correlated with infiltration of TAMs, polarization status of infiltrated TAMs, trastuzumab resistance and survival outcomes of GC patients. On exposure to CCL2, TAMs decreased the M1-like phenotype, thereby eliciting the trastuzumab resistance. CCL2 activated the transcription of ZC3H12A, which increased K63-linked deubiquitination and K48-linked auto-ubiquitination of TRAF6/3 to inactivate NF-κB signaling in TAMs. CD40 ×HER2 bsAb, which targeted the CD40 to restore the ubiquitination level of TRAF6/3, increased the M1-like phenotypic transformation of TAMs, and overcame trastuzumab resistance without immune-related adversary effects (irAEs). Conclusions We revealed a novel mechanism of trastuzumab resistance in HER2-positive GC via the CCL2-ZC3H12A-TRAF6/3 signaling axis, and presented a CD40 ×HER2 bsAb which showed great antitumor efficacy with few irAEs.
Collapse
Affiliation(s)
- Weilin Sun
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xi Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Daohan Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Lu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hai Lin
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhaoxiong Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yangpu Jia
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinyang Nie
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Tong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Weihua Fu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
9
|
Miyata Y, Matsuo T, Nakamura Y, Mitsunari K, Ohba K, Sakai H. Pathological Significance of Macrophages in Erectile Dysfunction Including Peyronie's Disease. Biomedicines 2021; 9:biomedicines9111658. [PMID: 34829887 PMCID: PMC8615952 DOI: 10.3390/biomedicines9111658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 12/17/2022] Open
Abstract
Erectile function is regulated by complex mechanisms centered on vascular- and nerve-related systems. Hence, dysregulation of these systems leads to erectile dysfunction (ED), which causes mental distress and decreases the quality of life of patients and their partners. At the molecular level, many factors, such as fibrosis, lipid metabolism abnormalities, the immune system, and stem cells, play crucial roles in the etiology and development of ED. Although phosphodiesterase type 5 (PDE5) inhibitors are currently the standard treatment agents for patients with ED, they are effective only in a subgroup of patients. Therefore, further insight into the pathological mechanism underlying ED is needed to discuss ED treatment strategies. In this review, we focused on the biological and pathological significance of macrophages in ED because the interaction of macrophages with ED-related mechanisms have not been well explored, despite their important roles in vasculogenic and neurogenic diseases. Furthermore, we examined the pathological significance of macrophages in Peyronie’s disease (PD), a cause of ED characterized by penile deformation (visible curvature) during erection and pain. Although microinjury and the subsequent abnormal healing process of the tunica albuginea are known to be important processes in this disease, the detailed etiology and pathophysiology of PD are not fully understood. This is the first review on the pathological role of macrophages in PD.
Collapse
Affiliation(s)
| | - Tomohiro Matsuo
- Correspondence: ; Tel.: +81-95-819-7340; Fax: +81-95-819-7343
| | | | | | | | | |
Collapse
|
10
|
Villegas G, Tar MT, Davies KP. Erectile dysfunction resulting from pelvic surgery is associated with changes in cavernosal gene expression indicative of cavernous nerve injury. Andrologia 2021; 54:e14247. [PMID: 34514620 DOI: 10.1111/and.14247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/30/2021] [Accepted: 09/02/2021] [Indexed: 10/20/2022] Open
Abstract
Pelvic surgery, even without direct cavernous nerve injury, carries a high risk of post-operative erectile dysfunction. The present studies were aimed at identifying molecular mechanisms by which pelvic surgery results in erectile dysfunction. As a model of pelvic surgery, male Sprague-Dawley rats underwent pelvic laparotomy, avoiding direct cavernous nerve injury. A second group of animals, serving as a model of direct cavernous nerve injury, underwent bilateral transection of the cavernous nerve. Cavernosometry demonstrated, that even in the absence of direct nerve injury, the pelvic surgery model exhibited significant erectile dysfunction 3 days post-operatively. Gene expression profiling also demonstrated that even in this animal model of nerve-sparing pelvic surgery, the profile of differentially expressed genes in cavernosal tissue was indicative of cavernous nerve injury. In addition, although 6 hr after surgery there were significant changes in circulating cytokine/chemokine levels, an inflammatory response in the major pelvic ganglion, cavernous nerve and cavernosal tissue was only observed 3 days post-surgery. Our results validate a rat model of pelvic surgery exhibiting erectile dysfunction and suggest systemic release of cytokines/chemokines following surgical trauma might mediate a pathological inflammatory response in tissues distal to the site of surgical trauma, indirectly resulting in cavernous nerve injury and erectile dysfunction.
Collapse
Affiliation(s)
- Guillermo Villegas
- Department of Urology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Moses Tarndie Tar
- Department of Urology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kelvin Paul Davies
- Department of Urology, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
11
|
Matsui H, Sopko NA, Campbell JD, Liu X, Reinhardt A, Weyne E, Castiglione F, Albersen M, Hannan JL, Bivalacqua TJ. Increased Level of Tumor Necrosis Factor-Alpha (TNF-α) Leads to Downregulation of Nitrergic Neurons Following Bilateral Cavernous Nerve Injury and Modulates Penile Smooth Tone. J Sex Med 2021; 18:1181-1190. [PMID: 37057424 DOI: 10.1016/j.jsxm.2021.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 04/07/2021] [Accepted: 05/03/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Erectile dysfunction (ED) after injury to peripheral cavernous nerve (CN) is partly a result of inflammation in pelvic ganglia, suggesting that ED may be prevented by inhibiting neuroinflammation. AIM The aim of this study is to examine temporal changes of TNF-α, after bilateral CN injury (BCNI), to evaluate effect of exogenous TNF-α on neurite outgrowth from major pelvic ganglion (MPG), and to investigate effect of TNF-α signal inhibition to evaluate effects of TNF-α on penile tone with TNF-α receptor knockout mice (TNFRKO). METHODS Seventy Sprague-Dawley rats were randomized to undergo BCNI or sham surgery. Sham rats' MPGs were harvested after 48 hours, whereas BCNI groups' MPGs were at 6, 12, 24, 48 hours, 7, or 14 days after surgery. qPCR was used to evaluate gene expression of markers for neuroinflammation in MPGs. Western blot was performed to evaluate TNF-α protein amount in MPGs. MPGs were harvested from healthy rats and cultured in Matrigel with TNF-α. Neurite outgrowth from MPGs was measured after 3 days, and TH and nNOS immunofluorescence was assessed. Wild type (WT) and TNFRKO mice were used to examine effect of TNF-α inhibition on smooth muscle function after BCNI. MPGs were harvested 48 hours after sham or BCNI surgery to evaluate gene expression of nNOS and TH. OUTCOMES Gene expression of TNF-α signaling pathway, Schwann cell and macrophage markers, protein expression of TNF-α in MPGs, and penile smooth muscle function to electrical field stimulation (EFS) were evaluated. RESULTS BCNI increased gene and protein expression of TNF-α in MPGs. Exogenous TNF-α inhibited MPG neurite outgrowth. MPGs cultured with TNF-α had decreased gene expression of nNOS (P < .05). MPGs cultured with TNF-α had shorter nNOS+ neurites than TH+ neurites (P < .01). Gene expression of nNOS was enhanced in TNFRKO mice compared to WT mice (P < .01). WT mice showed enhanced smooth muscle contraction of penises of WT mice was enhanced to EFS, compared to TNFKO (P < .01). Penile smooth-muscle relaxation to EFS was greater in TNFKO mice compared to WT (P < .01). CLINICAL TRANSLATION TNF-α inhibition may prevent ED after prostatectomy. STRENGTH/LIMITATIONS TNF-α inhibition might prevent loss of nitrergic nerve apoptosis after BCNI and preserve corporal smooth muscle function but further investigation is required to evaluate protein expression of nNOS in MPGs of TNFKO mice. CONCLUSIONS TNF-α inhibited neurite outgrowth from MPGs by downregulating gene expression of nNOS and TNFRKO mice showed enhanced gene expression of nNOS and enhanced penile smooth-muscle relaxation. Matsui H, Sopko NA, Campbell JD, et al. Increased Level of Tumor Necrosis Factor-Alpha (TNF-α) Leads to Downregulation of Nitrergic Neurons Following Bilateral Cavernous Nerve Injury and Modulates Penile Smooth Tone. J Sex Med 2021;18:1181-1190.
Collapse
Affiliation(s)
- Hotaka Matsui
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Urology, The University of Tokyo, Tokyo, Japan; Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Nikolai A Sopko
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jeffrey D Campbell
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Xiaopu Liu
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Emmanuel Weyne
- Laboratory for Experimental Urology, Department of Development and Regeneration, University of Leuven, Leuven, Belgium
| | - Fabio Castiglione
- Division of Surgery and Interventional Science, University College London, London, UK; Division of Oncology / Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Maarten Albersen
- Laboratory for Experimental Urology, Department of Development and Regeneration, University of Leuven, Leuven, Belgium
| | - Johanna L Hannan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Trinity J Bivalacqua
- The James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Luo Q, Liu R, Qu K, Liu G, Hang M, Chen G, Xu L, Jin Q, Guo D, Kang Q. Cangrelor ameliorates CLP-induced pulmonary injury in sepsis by inhibiting GPR17. Eur J Med Res 2021; 26:70. [PMID: 34229761 PMCID: PMC8262027 DOI: 10.1186/s40001-021-00536-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Sepsis is a common complication of severe wound injury and infection, with a very high mortality rate. The P2Y12 receptor inhibitor, cangrelor, is an antagonist anti-platelet drug. METHODS In our study, we investigated the protective mechanisms of cangrelor in CLP-induced pulmonary injury in sepsis, using C57BL/6 mouse models. RESULTS TdT-mediated dUTP Nick-End Labeling (TUNEL) and Masson staining showed that apoptosis and fibrosis in lungs were alleviated by cangrelor treatment. Cangrelor significantly promoted surface expression of CD40L on platelets and inhibited CLP-induced neutrophils in Bronchoalveolar lavage fluid (BALF) (p < 0.001). We also found that cangrelor decreased the inflammatory response in the CLP mouse model and inhibited the expression of inflammatory cytokines, IL-1β (p < 0.01), IL-6 (p < 0.05), and TNF-α (p < 0.001). Western blotting and RT-PCR showed that cangrelor inhibited the increased levels of G-protein-coupled receptor 17 (GPR17) induced by CLP (p < 0.001). CONCLUSION Our study indicated that cangrelor repressed the levels of GPR17, followed by a decrease in the inflammatory response and a rise of neutrophils in BALF, potentially reversing CLP-mediated pulmonary injury during sepsis.
Collapse
Affiliation(s)
- Qiancheng Luo
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Rui Liu
- Department of Endocrinology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Kaili Qu
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, 200135, People's Republic of China
| | - Guorong Liu
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Min Hang
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Guo Chen
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Lei Xu
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Qinqin Jin
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Dongfeng Guo
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China.
| | - Qi Kang
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China.
| |
Collapse
|
13
|
Wu J, Chen Z, Zhong F, Yang W, Ouyang X, Ma X, Zheng S, Wei H. Transplantation of Human Gingiva-Derived Mesenchymal Stem Cells Ameliorates Neurotic Erectile Dysfunction in a Rat Model. Front Bioeng Biotechnol 2021; 9:630076. [PMID: 34235136 PMCID: PMC8255925 DOI: 10.3389/fbioe.2021.630076] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/22/2021] [Indexed: 12/23/2022] Open
Abstract
Cavernous nerve injury (CNI) is the main cause of erectile dysfunction (ED) following pelvic surgery. Our previous studies have demonstrated that transplantation of different sources of mesenchymal stem cells (MSCs) was able to alleviate ED induced by CNI in rat models. However, little is known about the therapeutic effects of human gingiva-derived MSCs (hGMSCs) in CNI ED rats. Herein, we injected the hGMSCs around the bilateral major pelvic ganglia (MPG) in a rat model of CNI and evaluated their efficacy. The results showed that treatment of hGMSCs could significantly promote the recovery of erectile function, enhance smooth muscle and endothelial content, restore neuronal nitric oxide synthase (nNOS) expression, and attenuate cell apoptosis in penile tissue. Moreover, penile fibrosis was significantly alleviated after hGMSC administration. In addition, potential mechanism exploration indicated that hGMSCs might exert its functions via skewed macrophage polarity from M1 toward M2 anti-inflammatory phenotype. In conclusion, this study found that transplantation of hGMSCs significantly improved CNI-related ED, which might provide new clues to evaluate their pre-clinical application.
Collapse
Affiliation(s)
- Juekun Wu
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zehong Chen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fuyan Zhong
- Central Laboratory, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wende Yang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xi Ouyang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaolei Ma
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Songguo Zheng
- Department of Internal Medicine, Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH, United States
| | - Hongbo Wei
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
Gu Y, Niu X, Yin L, Wang Y, Yang Y, Yang X, Zhang Q, Ji H. Enhancing Fatty Acid Catabolism of Macrophages Within Aberrant Breast Cancer Tumor Microenvironment Can Re-establish Antitumor Function. Front Cell Dev Biol 2021; 9:665869. [PMID: 33937269 PMCID: PMC8081981 DOI: 10.3389/fcell.2021.665869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/25/2021] [Indexed: 12/21/2022] Open
Abstract
Triple-negative breast cancer (TNBC) remains an intractable challenge owing to its aggressive nature and lack of any known therapeutic targets. Macrophages play a crucial role in cancer promotion and poor prognosis within the tumor microenvironment (TME). The phagocytosis checkpoint in macrophages has broader implications for current cancer immunotherapeutic strategies. Here, we demonstrate the modulation in the antitumor activity of macrophages within the aberrant metabolic microenvironment of TNBC by metabolic intervention. The co-culture of macrophages with TNBC cell lines led to a decrease in both their phagocytic function and expression of interleukin (IL)-1β and inducible nitric oxide synthase (iNOS). The transcription of glycolysis and fatty acid (FA) catabolism-related factors was inhibited within the dysregulated tumor metabolic microenvironment. Enhancement of FA catabolism by treatment with the peroxisome proliferator-activated receptor-alpha (PPAR-α) agonist, fenofibrate (FF), could re-establish macrophages to gain their antineoplastic activity by activating the signal transducer and activator of transcription 1 (STAT1) signaling pathway and increasing ATP production by FA oxidation. The combination of fenofibrate and anti-CD47 therapy significantly inhibited tumor growth in a 4T1 tumor-bearing mouse model. In conclusion, the enhancement of FA catabolism of macrophages could re-establish them to resume antitumor activity in the TME. Anti-CD47 therapy combined with fenofibrate may serve as a novel and potential immunotherapeutic approach for the treatment of TNBC.
Collapse
Affiliation(s)
- Yucui Gu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Xingjian Niu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Lei Yin
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Yiran Wang
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Yue Yang
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Xudong Yang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Qingyuan Zhang
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Hongfei Ji
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Sciences, Harbin, China
| |
Collapse
|
15
|
Chang Q, Hao Y, Wang Y, Zhou Y, Zhuo H, Zhao G. Bone marrow mesenchymal stem cell-derived exosomal microRNA-125a promotes M2 macrophage polarization in spinal cord injury by downregulating IRF5. Brain Res Bull 2021; 170:199-210. [PMID: 33609602 DOI: 10.1016/j.brainresbull.2021.02.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/03/2021] [Accepted: 02/10/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Spinal cord injury (SCI) may cause loss of locomotor function, and macrophage is a major cell type in response to SCI with M1- and M2-phenotypes. The protective role of bone marrow mesenchymal stem cells (BMMSC)-derived exosomes (B-Exo) in SCI has been underscored, while their regulation on M2 macrophage polarization and the mechanism remain to be clarified. METHODS A rat model of SCI was developed and treated with extracted B-Exo. Recovery of motor function was assessed by Basso-Beattie-Bresnahan (BBB) score. The apoptosis and degeneration of neurons, and macrophage polarization were evaluated. Subsequently, genes differentially expressed in the rat spinal cord after B-Exo treatment were analyzed. Later, the relationships between B-Exo and interferon regulatory factor 5 (IRF5) or macrophage polarization were clarified. Later, the upstream microRNAs (miRNAs) of IRF5 were validated by bioinformatics prediction and dual-luciferase experiments. Finally, the role of miR-125a in the neuroprotection of SCI was verified by rescue experiments. RESULTS B-Exo promoted the recovery of locomotor function and M2-phenotype polarization, whereas inhibited neuronal apoptosis and degeneration and the inflammatory response caused by SCI in rats. In addition, IRF5 expression was reduced after B-Exo treatment. IRF5 promoted macrophage polarization towards M1-phenotype and secretion of inflammatory factors. There is a binding relationship between miR-125a and IRF5. Knockdown of miR-125a in B-Exo increased IRF5 expression in spinal cord tissues of SCI rats and attenuated the neuroprotective effect of B-Exo against SCI. CONCLUSION Exosomal miR-125a derived from BMMSC exerts neuroprotective effects by targeting and negatively regulating IRF5 expression in SCI rats.
Collapse
Affiliation(s)
- Qing Chang
- Department Two of Spine, Luoyang Orthopedic-Traumatological Hospital of Henan Province (Henan Provincial Orthopedic Hospital), Luoyang, 471000, Henan, PR China
| | - Yupeng Hao
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, PR China
| | - Yifan Wang
- Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, PR China
| | - Yingjie Zhou
- Department Two of Spine, Luoyang Orthopedic-Traumatological Hospital of Henan Province (Henan Provincial Orthopedic Hospital), Luoyang, 471000, Henan, PR China
| | - Hanjie Zhuo
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, PR China
| | - Gang Zhao
- Department Two of Spine, Luoyang Orthopedic-Traumatological Hospital of Henan Province (Henan Provincial Orthopedic Hospital), Luoyang, 471000, Henan, PR China.
| |
Collapse
|
16
|
Kaya-Sezginer E, Gur S. The Inflammation Network in the Pathogenesis of Erectile Dysfunction: Attractive Potential Therapeutic Targets. Curr Pharm Des 2021; 26:3955-3972. [PMID: 32329680 DOI: 10.2174/1381612826666200424161018] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 03/17/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Erectile dysfunction (ED) is an evolving health problem in the aging male population. Chronic low-grade inflammation is a critical component of ED pathogenesis and a probable intermediate stage of endothelial dysfunction, especially in metabolic diseases, with the inclusion of obesity, metabolic syndrome, and diabetes. OBJECTIVE This review will present an overview of preclinical and clinical data regarding common inflammatory mechanisms involved in the pathogenesis of ED associated with metabolic diseases and the effect of antiinflammatory drugs on ED. METHODS A literature search of existing pre-clinical and clinical studies was performed on databases [Pubmed (MEDLINE), Scopus, and Embase] from January 2000 to October 2019. RESULTS Low-grade inflammation is a possible pathological role in endothelial dysfunction as a consequence of ED and other related metabolic diseases. Increased inflammation and endothelial/prothrombotic markers can be associated with the presence and degree of ED. Pharmacological therapy and modification of lifestyle and risk factors may have a significant role in the recovery of erectile response through reduction of inflammatory marker levels. CONCLUSION Inflammation is the least common denominator in the pathology of ED and metabolic disorders. The inflammatory process of ED includes a shift in the complex interactions of cytokines, chemokines, and adhesion molecules. These data have established that anti-inflammatory agents could be used as a therapeutic opportunity in the prevention and treatment of ED. Further research on inflammation-related mechanisms underlying ED and the effect of therapeutic strategies aimed at reducing inflammation is required for a better understanding of the pathogenesis and successful management of ED.
Collapse
Affiliation(s)
- Ecem Kaya-Sezginer
- Department of Biochemistry and Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Serap Gur
- Department of Biochemistry and Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
17
|
European Society for Sexual Medicine Consensus Statement on the Use of the Cavernous Nerve Injury Rodent Model to Study Postradical Prostatectomy Erectile Dysfunction. Sex Med 2020; 8:327-337. [PMID: 32674971 PMCID: PMC7471127 DOI: 10.1016/j.esxm.2020.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/16/2020] [Accepted: 06/14/2020] [Indexed: 01/24/2023] Open
Abstract
INTRODUCTION Rodent animal models are currently the most used in vivo model in translational studies looking into the pathophysiology of erectile dysfunction after nerve-sparing radical prostatectomy. AIM This European Society for Sexual Medicine (ESSM) statement aims to guide scientists toward utilization of the rodent model in an appropriate, timely, and proficient fashion. METHODS MEDLINE and EMBASE databases were searched for basic science studies, using a rodent animal model, looking into the consequence of pelvic nerve injury on erectile function. MAIN OUTCOME MEASURES The authors present a consensus on how to best perform experiments with this rodent model, the details of the technique, and highlight possible pitfalls. RESULTS Owing to the specific issue-basic science-Oxford 2011 Levels of Evidence criteria cannot be applied. However, ESSM statements on this topic will be provided in which we summarize the ESSM position on various aspects of the model such as the use of the Animal Research Reporting In Vivo Experiments guideline and the of common range parameter for nerve stimulation. We also highlighted the translational limits of the model. CONCLUSION The following statements were formulated as a suggestive guidance for scientists using the cavernous nerve injury model. With this, we hope to standardize and further improve the quality of research in this field. It must be noted that this model has its limitations. Weyne E, Ilg MM, Cakir OO, et al. European Society for Sexual Medicine Consensus Statement on the Use of the Cavernous Nerve Injury Rodent Model to Study Postradical Prostatectomy Erectile Dysfunction. Sex Med 2020;8:327-337.
Collapse
|
18
|
Fang H, Yang M, Pan Q, Jin HL, Li HF, Wang RR, Wang QY, Zhang JP. MicroRNA-22-3p alleviates spinal cord ischemia/reperfusion injury by modulating M2 macrophage polarization via IRF5. J Neurochem 2020; 156:106-120. [PMID: 32406529 DOI: 10.1111/jnc.15042] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 04/17/2020] [Accepted: 05/06/2020] [Indexed: 02/05/2023]
Abstract
Cell death after spinal cord ischemia/reperfusion (I/R) can occur through necrosis, apoptosis, and autophagy, resulting in changes to the immune environment. However, the molecular mechanism of this immune regulation is not clear. Accumulating evidence indicates that microRNAs (miRs) play a crucial role in the pathogenesis of spinal cord I/R injury. Here, we hypothesized miR-22-3p may be involved in spinal cord I/R injury by interacting with interferon regulatory factor (IRF) 5. Rat models of spinal cord I/R injury were established by 12-min occlusion of the aortic arch followed by 48-hr reperfusion, with L4-6 segments of spinal cord tissues collected. MiR-22-3p agomir, a lentivirus-delivered siRNA specific for IRF5, or a lentivirus expressing wild-type IRF5 was injected intrathecally to rats with I/R injury to evaluate the effects of miR-22-3p and IRF5 on hindlimb motor function. Macrophages isolated from rats were treated with miR-22-3p mimic or siRNA specific for IRF5 to evaluate their effects on macrophage polarization. The levels of IL-1β and TNF-α in spinal cord tissues were detected by ELISA. miR-22-3p was down-regulated, whereas IRF5 was up-regulated in rat spinal cord tissues following I/R. IRF5 was a target gene of miR-22-3p and could be negatively regulated by miR-22-3p. Silencing IRF5 or over-expressing miR-22-3p relieved inflammation, elevated Tarlov score, and reduced the degree of severity of spinal cord I/R injury. Increased miR-22-3p facilitated M2 polarization of macrophages and inhibited inflammation in tissues by inhibiting IRF5, thereby attenuating spinal cord I/R injury. Taken together, these results demonstrate that increased miR-22-3p can inhibit the progression of spinal cord I/R injury by repressing IRF5 in macrophages, highlighting the discovery of a promising new target for spinal cord I/R injury treatment.
Collapse
Affiliation(s)
- Hua Fang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, P.R. China.,Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, P.R. China.,Laboratory of Anesthesiology and Perioperative Medicine, Guizhou University School of Medicine, Guiyang, P.R. China
| | - Miao Yang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, P.R. China.,Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, P.R. China.,Laboratory of Anesthesiology and Perioperative Medicine, Guizhou University School of Medicine, Guiyang, P.R. China
| | - Qin Pan
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, P.R. China.,Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, P.R. China.,Laboratory of Anesthesiology and Perioperative Medicine, Guizhou University School of Medicine, Guiyang, P.R. China
| | - Hon-Ling Jin
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, P.R. China.,Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, P.R. China.,Laboratory of Anesthesiology and Perioperative Medicine, Guizhou University School of Medicine, Guiyang, P.R. China
| | - Hua-Feng Li
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, P.R. China
| | - Ru-Rong Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Quan-Yun Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Jian-Ping Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, P.R. China.,Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, P.R. China.,Laboratory of Anesthesiology and Perioperative Medicine, Guizhou University School of Medicine, Guiyang, P.R. China
| |
Collapse
|
19
|
CCL2 promotes macrophages-associated chemoresistance via MCPIP1 dual catalytic activities in multiple myeloma. Cell Death Dis 2019; 10:781. [PMID: 31611552 PMCID: PMC6791869 DOI: 10.1038/s41419-019-2012-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/27/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
We previously showed that the chemokine CCL2 can recruit macrophages (Mφs) to the bone marrow (BM) in multiple myeloma (MM) and that myeloma-associated Mφs are important in drug resistance. Here, we explore the role of increased CCL2 expression in the BM microenvironment of MM and elucidate the underlying mechanism. Our results show that CCL2 expression is associated with the treatment status of MM patients. Mφs interact with MM cells and further upregulate their expression of CCL2. These increased level of CCL2 polarizes Mφs toward the M2-like phenotype and promotes Mφs to protect MM cells from drug-induced apoptosis. Mechanistically, CCL2 upregulated the expression of the immunosuppressive molecular MCP-1-induced protein (MCPIP1) in Mφs. MCPIP1 mediates Mφs’ polarization and protection via dual catalytic activities. Additionally, we found that CCL2 induces MCPIP1 expression via the JAK2-STAT3 signaling pathway. Taken together, our results indicate that increased CCL2 expression in MM patients’ BM polarizes Mφs toward the M2-like phenotype and promotes the protective effect of Mφs through MCPIP1, providing novel insight into the mechanism of Mφs-mediated drug resistance in MM.
Collapse
|
20
|
Calmasini FB, Klee N, Webb RC, Priviero F. Impact of Immune System Activation and Vascular Impairment on Male and Female Sexual Dysfunction. Sex Med Rev 2019; 7:604-613. [PMID: 31326360 DOI: 10.1016/j.sxmr.2019.05.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/03/2019] [Accepted: 05/30/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Male and female sexual dysfunction (SD) is considered a multifactorial condition. Numerous studies have shown the involvement of inflammatory processes in this pathological condition. Sexual intercourse requires healthy and functioning vessels to supply the pelvic region in both males and females, generating penile erection and clitoral and vaginal lubrication, respectively. Cardiovascular diseases and associated risk factors may contribute negatively to pelvic blood flow, possibly through immune system activation. AIM The study aimed to address the correlation between vascular inflammation driven by immune system activation and SD in males and females. METHODS A literature review was performed to identify articles addressing male and female SD and vascular inflammation. Key words included "male and female sexual dysfunction," "vascular inflammation," "iliac and pudendal arteries dysfunction," "genitourinary tract," and "blood flow." MAIN OUTCOME MEASURES Management of systemic and local inflammation may be a useful alternative to improve SD and reduce the risk of cardiovascular diseases in the future. RESULTS Increased levels of cytokines and chemokines have been detected in humans and animals with hypertension, obesity, and diabetic conditions. Chronic activation of the innate immune system, especially by pathogen- or damage-associated molecular patterns, and metabolic-related disorders may act as triggers further contributing to an increased inflammatory condition. Due to the reduced size of vessels, SD and retinal vascular impairments have been shown to be predictive factors for cardiovascular diseases. Therefore, considering that blood flow to the genitalia is essential for sexual function, endothelial dysfunction and vascular remodeling, secondary to chronic immune system activation, may be implicated in male and female vasculogenic SD. CONCLUSIONS Several conditions appear to play a role in SD. In the present review, we have identified a role for the immune system in generating vascular and tissue impairments contributing to erectile dysfunction and female SD. Calmasini FB, Klee N, Webb RC, et al. Impact of Immune System Activation and Vascular Impairment on Male and Female Sexual Dysfunction. Sex Med Rev 2019;7:604-613.
Collapse
Affiliation(s)
- Fabiano B Calmasini
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Deparment of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil.
| | - Nicole Klee
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - R Clinton Webb
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Fernanda Priviero
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
21
|
Wang LX, Zhang SX, Wu HJ, Rong XL, Guo J. M2b macrophage polarization and its roles in diseases. J Leukoc Biol 2018; 106:345-358. [PMID: 30576000 PMCID: PMC7379745 DOI: 10.1002/jlb.3ru1018-378rr] [Citation(s) in RCA: 498] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/07/2018] [Accepted: 12/09/2018] [Indexed: 12/14/2022] Open
Abstract
Macrophages play an important role in a wide variety of physiologic and pathologic processes. Plasticity and functional polarization are hallmarks of macrophages. Macrophages commonly exist in two distinct subsets: classically activated macrophages (M1) and alternatively activated macrophages (M2). M2b, a subtype of M2 macrophages, has attracted increasing attention over the past decade due to its strong immune‐regulated and anti‐inflammatory effects. A wide variety of stimuli and multiple factors modulate M2b macrophage polarization in vitro and in vivo. M2b macrophages possess both protective and pathogenic roles in various diseases. Understanding the mechanisms of M2b macrophage activation and the modulation of their polarization might provide a great perspective for the design of novel therapeutic strategies. The purpose of this review is to discuss current knowledge of M2b macrophage polarization, the roles of M2b macrophages in a variety of diseases and the stimuli to modulate M2b macrophage polarization.
Collapse
Affiliation(s)
- Le-Xun Wang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Joint Laboratory of Guangdong, Hong Kong and Macao on Glycolipid Metabolic Diseases, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Sheng-Xi Zhang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Joint Laboratory of Guangdong, Hong Kong and Macao on Glycolipid Metabolic Diseases, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hui-Juan Wu
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Joint Laboratory of Guangdong, Hong Kong and Macao on Glycolipid Metabolic Diseases, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiang-Lu Rong
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Joint Laboratory of Guangdong, Hong Kong and Macao on Glycolipid Metabolic Diseases, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Joint Laboratory of Guangdong, Hong Kong and Macao on Glycolipid Metabolic Diseases, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
22
|
Zhan TW, Tian YX, Wang Q, Wu ZX, Zhang WP, Lu YB, Wu M. Cangrelor alleviates pulmonary fibrosis by inhibiting GPR17-mediated inflammation in mice. Int Immunopharmacol 2018; 62:261-269. [PMID: 30036769 DOI: 10.1016/j.intimp.2018.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/07/2018] [Accepted: 06/04/2018] [Indexed: 12/19/2022]
Abstract
Pulmonary fibrosis is a progressive and intractable lung disease. Macrophages play a critical role in the progression of pulmonary fibrosis. Cangrelor, an anti-platelet agent, is also a non-selective Gprotein-coupled receptor 17 (GPR17) antagonist. GPR17 mediates microglial inflammation in the chronic phase of cerebral ischemia and regulates allergic pulmonary inflammation. In this study, we observed the effects of cangrelor on bleomycin (BLM)-induced macrophage cellular inflammation and BLM-induced pulmonary fibrosis in C57BL/6J mice. We found that BLM significantly increased GPR17 expression, the mRNA synthesis and release of inflammatory cytokines including TNF-α, IL-6 and TGF-β1 in murine RAW 264.7 macrophage cells. Knockdown of GPR17 attenuated the BLM-induced inflammatory responses. Cangrelor (2.5 μM-10 μM) significantly alleviated BLM-induced inflammatory response in RAW 264.7 macrophage cells in concentration-dependent manner. In BLM-induced fibrotic mouse lungs, GPR17 expression and GPR17-positive macrophages were increased. Cangrelor (2.5 mg/kg-10 mg/kg) alleviated pulmonary fibrosis in dose-dependent manner. Cangrelor not only reduced the number of GPR17-positive macrophages, but also decreased BLM-induced mRNA synthesis and release of inflammatory cytokine. As such, we concluded that cangrelor alleviates BLM-induced pulmonary fibrosis by suppressing GPR17-mediated inflammation. Cangrelor could be a potential therapeutic drug for pulmonary fibrosis.
Collapse
Affiliation(s)
- Tian-Wei Zhan
- Department of Thoracic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jie-Fang Road, Hangzhou, Zhejiang 310009, China
| | - Yu-Xin Tian
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yu-Hang-Tang Road, Hangzhou, Zhejiang 310058, China
| | - Qi Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jie-Fang Road, Hangzhou, Zhejiang 310009, China
| | - Zi-Xiang Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jie-Fang Road, Hangzhou, Zhejiang 310009, China
| | - Wei-Ping Zhang
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yu-Hang-Tang Road, Hangzhou, Zhejiang 310058, China
| | - Yun-Bi Lu
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yu-Hang-Tang Road, Hangzhou, Zhejiang 310058, China
| | - Ming Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jie-Fang Road, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
23
|
Yoshida T, Sopko NA, Kates M, Liu X, Joice G, McConkey DJ, Bivalacqua TJ. Three-dimensional organoid culture reveals involvement of Wnt/β-catenin pathway in proliferation of bladder cancer cells. Oncotarget 2018; 9:11060-11070. [PMID: 29541396 PMCID: PMC5834271 DOI: 10.18632/oncotarget.24308] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 01/19/2018] [Indexed: 12/23/2022] Open
Abstract
There has been increasing awareness of the importance of three-dimensional culture of cancer cells. Tumor cells growing as multicellular spheroids in three-dimensional culture, alternatively called organoids, are widely believed to more closely mimic solid tumors in situ. Previous studies concluded that the Wnt/β-catenin pathway is required for regeneration of the normal urothelium after injury and that β-catenin is upregulated in human bladder cancers, but no clear evidence has been advanced to support the idea that the Wnt/β-catenin pathway is directly involved in deregulated proliferation and the other malignant characteristics of bladder cancer cells. Here we report that the Wnt/β-catenin pathway activator, CHIR99021, promoted proliferation of established human bladder cancer cell lines when they were grown in organoid culture but not when they were grown in conventional adherent cultures. CHIR99021 activated Wnt/β-catenin pathway in bladder cancer cell lines in organoid culture. CHIR99021 also stimulated proliferation and the Wnt/b-catenin pathway in primary human bladder cancer organoids. RNAi-mediated knockdown of β-catenin blocked growth of organoids. The effects of CHIR99021 were associated with decreased expression of the urothelial terminal differentiation marker, cytokeratin 20. Our data suggest that the Wnt/β-catenin pathway is required for the proliferation of bladder cancer cells in three-dimensional organoid culture and provide a concrete example of why organoid culture is important for cancer research.
Collapse
Affiliation(s)
- Takahiro Yoshida
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Nikolai A. Sopko
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Max Kates
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Xiaopu Liu
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Gregory Joice
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - David J. McConkey
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- The Johns Hopkins Greenberg Bladder Cancer Institute, Baltimore, Maryland, USA
| | - Trinity J. Bivalacqua
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- The Johns Hopkins Greenberg Bladder Cancer Institute, Baltimore, Maryland, USA
| |
Collapse
|