1
|
Majidpour M, Azizi SG, Davodabadi F, Sabeti Akbar-Abad M, Abdollahi Z, Sargazi S, Shahriari H. Recent advances in TGF-β signaling pathway in COVID-19 pathogenesis: A review. Microb Pathog 2025; 199:107236. [PMID: 39701478 DOI: 10.1016/j.micpath.2024.107236] [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: 09/02/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/21/2024]
Abstract
The coronavirus disease 2019 (COVID-19) has resulted in approximately 7.0 million fatalities between 2019 and 2022, underscoring a pressing need for comprehensive research into its underlying mechanisms and therapeutic avenues. A distinctive feature of severe COVID-19 is the dysregulated immune response characterized by excessive activation of immune cells and the consequent cytokine storms. Recent advancements in our understanding of cellular signaling pathways have illuminated the role of Transforming Growth Factor Beta (TGF-β) as a pivotal signaling molecule with significant implications for the pathogenesis of infectious diseases, including COVID-19. Emerging evidence reveals that TGF-β signaling, when activated by viral components or secondary pathways, adversely affects diverse cell types, particularly immune cells, and lung tissue, leading to complications such as pulmonary fibrosis. In our review article, we critically evaluate recent literature on the involvement of TGF-β signaling in the progression of COVID-19. We discuss a range of pharmacological interventions, including nintedanib, pirfenidone, corticosteroids, proton pump inhibitors, and histone deacetylase inhibitors, and their potential to modulate the TGF-β pathway in the context of COVID-19 treatment. Additionally, we explore ongoing clinical trials involving mesenchymal stem cells, low-dose radiation therapy, and artemisinin derivatives to assess their impact on TGF-β levels and subsequent clinical outcomes in COVID-19 patients. This review is particularly relevant at this juncture as the global health community continues to grapple with the ramifications of the COVID-19 pandemic, highlighting the urgent need for targeted therapeutic strategies aimed at TGF-β modulation to mitigate disease severity and improve patient outcomes.
Collapse
Affiliation(s)
- Mahdi Majidpour
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Seyed Ghader Azizi
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Fatemeh Davodabadi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahboobeh Sabeti Akbar-Abad
- Department of Clinical Biochemistry, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Zahra Abdollahi
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran.
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Hossein Shahriari
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| |
Collapse
|
2
|
Phuong NQ, Bilal M, Nawaz A, Anh LD, Memoona, Aslam MR, Khalid S, Kado T, Watanabe Y, Nishimura A, Igarashi Y, Okabe K, Hirabayashi K, Yamamoto S, Nakagawa T, Mori H, Usui I, Fujisaka S, Hayashi R, Tobe K. Role of transforming growth factor-β1 in regulating adipocyte progenitors. Sci Rep 2025; 15:941. [PMID: 39824986 DOI: 10.1038/s41598-024-81917-7] [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: 07/02/2024] [Accepted: 11/29/2024] [Indexed: 01/20/2025] Open
Abstract
Adipose tissue (AT) metabolism involves coordinating various cells and cellular processes to regulate energy storage, release, and overall metabolic homeostasis. Therein, macrophage and its cytokine are important in controlling tissue homeostasis. Among cytokines, the role of transforming growth factor-β1 (Tgf-β1), a cytokine abundantly expressed in CD206+ M2-like macrophage and correlated with the expansion of AT and fibrosis, in AT metabolism, remains unknown. We used CD206CreERT2; Tgf-β1f/f mouse model in which the Tgf-β1 gene was conditionally deleted in CD206+ M2-like macrophages followed by tamoxifen administration, to investigate the role of the Tgf-β1 gene in glucose and insulin metabolism. Our data demonstrated that lack of CD206+ M2-like macrophages derived Tgf-β1 gene improved glucose metabolism and insulin sensitivity by enhancing adipogenesis via hyperplasia. The Tgf-β1 gene, specifically from CD206+ M2-like macrophages, deletion stimulated APs' proliferation and differentiation, leading to the generation of smaller mature adipocytes, therefore enhancing insulin sensitivity and improving glucose metabolism under normal chow conditions. Our study brings a new perspective that Tgf-β1 gene deletion specific from CD206+ M2-like macrophage promotes adipocyte hyperplasia, improving glucose homeostasis and insulin sensitivity in the lean state.
Collapse
Affiliation(s)
- Nguyen Quynh Phuong
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
- Clinical Oncology, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Muhammad Bilal
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
- Research Center for Pre-Disease Science, Faculty of Education and Research Promotion, University of Toyama, Toyama, 930-0194, Japan.
- Advanced Postdoctoral Fellowships of the Japan Diabetes Society (JDS), Tokyo, Japan.
| | - Allah Nawaz
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Le Duc Anh
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Memoona
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Muhammad Rahil Aslam
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Sana Khalid
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Tomonobu Kado
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Yoshiyuki Watanabe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Ayumi Nishimura
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
- Faculty of Education and Research Promotion, University of Toyama, Toyama, 930-0194, Japan
| | - Yoshiko Igarashi
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
- JSPS Research Fellowship for Young Scientist Japan, Tokyo, Japan
| | - Keisuke Okabe
- Toyama University Hospital, Center for Clinical and Translational Research, Toyama, Japan
| | - Kenichi Hirabayashi
- Department of Diagnostic Pathology, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Seiji Yamamoto
- Department of Pathology, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Isao Usui
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Tochigi, Japan
| | - Shiho Fujisaka
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Ryuji Hayashi
- Clinical Oncology, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Kazuyuki Tobe
- Research Center for Pre-Disease Science, Faculty of Education and Research Promotion, University of Toyama, Toyama, 930-0194, Japan.
- Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|
3
|
Dietsche L, Stirm K, Lysenko V, Schneidawind C, Tzankov A, Müller A, Theocharides APA. Loss of SMAD1 in acute myeloid leukemia with KMT2A::AFF1 and KMT2A::MLLT3 fusion genes. Front Oncol 2025; 14:1481713. [PMID: 39834944 PMCID: PMC11743462 DOI: 10.3389/fonc.2024.1481713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction KMT2A-rearrangements define a subclass of acute leukemias characterized by a distinct gene expression signature linked to the dysfunctional oncogenic fusion proteins arising from various chromosomal translocations involving the KMT2A (also known as MLL1) gene. Research on the disease pathomechanism in KMT2A-rearranged acute leukemias has mainly focused on the upregulation of the stemness-related genes of the HOX-family and their co-factor MEIS1. Results Here we report the KMT2A::AFF1 and KMT2A::MLLT3 fusion gene-dependent downregulation of SMAD1, a TGF-β signaling axis transcription factor. SMAD1 expression is lost in the majority of AML patient samples and cell lines containing the two fusion genes KMT2A::AFF1 and KMT2A::MLLT3 compared to non-rearranged controls. Loss of SMAD1 expression is inducible by introducing the respective two KMT2A fusion genes into hematopoietic stem and progenitor cells. The loss of SMAD1 correlated with a markedly reduced amount of H3K4me3 levels at the SMAD1 promoter in tested cells with KMT2A::AFF1 and KMT2A::MLLT3. The expression of SMAD1 in cells with KMT2A::AFF1 fusion genes impacted the growth of cells in vitro and influenced engraftment of the KMT2A::AFF1 cell line MV4-11 in vivo. In MV4-11 cells SMAD1 expression caused a downregulation of HOXA9 and MEIS1, which was reinforced by TGF-β stimulation. Moreover, in MV4-11 cells SMAD1 presence sensitized cells for TGF-β mediated G1-arrest. Conclusion Overall, our data contributes to the understanding of the role of TGF-β signaling in acute myeloid leukemia with KMT2A::AFF1 by showing that SMAD1 loss can influence the growth dynamics and contribute to the pathogenic expression of disease driving factors.
Collapse
Affiliation(s)
- Lisa Dietsche
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Kristin Stirm
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Veronika Lysenko
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Corina Schneidawind
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Alexandar Tzankov
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Alexandre P. A. Theocharides
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
Yang L, Geng L, Zhang X, Lu J, Zhang H, Wu G, Huang S. Interaction of AURKA with TRIM28 revives dormant LSCC cells via Akt signaling pathway to promote LSCC metastasis. Cancer Cell Int 2025; 25:2. [PMID: 39754170 PMCID: PMC11697802 DOI: 10.1186/s12935-024-03620-x] [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: 06/23/2024] [Accepted: 12/14/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Specific molecular mechanisms by which AURKA promoted LSCC metastasis were still unknown. METHODS Bioinformatic analysis was performed the relationship between TRIM28 and LSCC. Immunohistochemistry, Co-IP assay, Rt-PCR and Western Blot were used to examine the expression of related molecular. Flow cytometry was used to examine cell numbers of G0/G1 phase. Plate colony formation, wound healing, migration, invasion and tail vein injection in nude mice assays were applied to examine the proliferation, movement, migration, invasion and metastasis of LSCC. RESULTS TRIM28 was significantly correlated with LSCC. TRIM28 highly expressed in LSCC and the high TRIM28 expression was related to TNM stage and poor clinical prognosis. Furthermore, AURKA could regulate TRIM28. In addition, deprivation TRIM28 expression induced LSCC cells into dormant state and inhibited LSCC metastasis. Akt signaling pathway played an essential role in promoting the tumor-promoting effects induced by TRIM28. CONCLUSION AURKA mediated TRIM28 to revive dormant LSCC cells via Akt signaling pathway to promote LSCC metastasis, targeting TRIM28 might provide a potential treatment strategy for LSCC.
Collapse
Affiliation(s)
- Liyun Yang
- Department of Otolaryngology, Pudong Gongli Hospital, Shanghai, 200135, China.
- Department of Otolaryngology, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou, 215600, China.
| | - Liang Geng
- Department of Otolaryngology, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou, 215600, China
| | - Xinxin Zhang
- Department of Otolaryngology, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou, 215600, China
| | - Jing Lu
- Department of Otolaryngology, Pudong Gongli Hospital, Shanghai, 200135, China
| | - Hao Zhang
- Department of Otolaryngology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Geping Wu
- Department of Otolaryngology, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou, 215600, China.
| | - Shuixian Huang
- Department of Otolaryngology, Pudong Gongli Hospital, Shanghai, 200135, China.
| |
Collapse
|
5
|
Qin H, Yuan M, Yuan Y, Xia F, Yang Y. NEK2 promotes colorectal cancer progression by activating the TGF-β/Smad2 signaling pathway. Transl Oncol 2025; 51:102186. [PMID: 39499996 PMCID: PMC11570754 DOI: 10.1016/j.tranon.2024.102186] [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: 09/23/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/21/2024] Open
Abstract
Colorectal cancer (CRC) is a prevalent malignancy with poor patient survival, and NIMA-associated kinase 2 (NEK2) has been implicated in the pathogenesis and progression of various cancers, including CRC. This study aimed to investigate the impact of NEK2 on CRC cell functionality and its interaction with the TGF-β/Smad signaling pathway. NEK2 expression in CRC tissues and cell lines was assessed, and its association with patient survival was analyzed. Functional assays, including NEK2 knockdown via lentiviral infection, RT-qPCR, Western blotting, CCK-8 assay, Transwell migration, invasion assays, and goblet cell formation assays, were employed to evaluate NEK2's effects on CRC cell proliferation, migration, invasion, and stemness. Mechanistic studies explored the TGF-β/Smad2 signaling pathway, utilizing co-immunoprecipitation (Co-IP) and protein interaction analyses. In vivo experiments further evaluated NEK2's role in tumor initiation, metastasis, and chemoresistance. NEK2 was found to be upregulated in CRC tissues and correlated with poor survival. NEK2 knockdown inhibited CRC cell behaviors, while NEK2 activated the TGF-β/Smad2 signaling pathway through Smad2/3 phosphorylation. Overexpression of Smad2/3 reversed NEK2 knockdown effects, confirming the importance of this pathway in CRC. In vivo, NEK2 promoted tumor initiation, metastasis, and chemoresistance, effects partially reversed by Smad2/3 overexpression. These findings reveal the critical role of NEK2 in CRC progression and underscore its potential as a therapeutic target, offering new insights into the molecular mechanisms driving CRC and informing targeted therapy development.
Collapse
Affiliation(s)
- Hai Qin
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, No. 206, Sixian Street, Baiyun District, Guiyang City, Guizhou Province, China.
| | - Manqin Yuan
- Department of Clinical Laboratory Medicine, Guizhou Medical University, No. 9, Beijing Road, Yunyan District, Guiyang City, Guizhou Province, China
| | - Yaqin Yuan
- Microbiological Laboratory, Guizhou Center for Medical Device Testing, No.247, South Section of Xintian Avenue, Yunyan District, Guiyang City, 550001, Guizhou Province, China
| | - Fengqiong Xia
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, No. 206, Sixian Street, Baiyun District, Guiyang City, Guizhou Province, China
| | - Yonghong Yang
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, No. 206, Sixian Street, Baiyun District, Guiyang City, Guizhou Province, China.
| |
Collapse
|
6
|
Li Y, Jiang H, Li X, Zhu H, Dai Y, Zhang J, Sun Y, Chu X, Ju W, Xu M, Li Z, Zeng L, Xu K, Qiao J. Platelet-Specific Deletion of TGF-β1 Impairs Septic Thrombosis in Mice-Brief Report. Arterioscler Thromb Vasc Biol 2025; 45:136-143. [PMID: 39633577 DOI: 10.1161/atvbaha.124.322029] [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: 10/20/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Sepsis is featured as a systemic inflammation and thrombosis induced by infection. TGF-β (transforming growth factor-β) 1 is mainly secreted from platelets and plays a role in immune response and inflammation. Whether platelet-derived TGF-β1 participates in sepsis remains unclear. This study intends to investigate its role in sepsis in mice. METHODS Platelet-specific TGF-β1 knockout mice received cecal ligation and puncture surgery to induce sepsis followed by the analysis of survival time, platelets number, pathology changes of lung and liver, liver function, the recruitment of platelets, neutrophils and monocytes, and neutrophil extracellular traps' formation. In addition, adoptive transfer of wild-type platelets into platelet-specific TGF-β1 knockout mice was performed to further evaluate the role of TGF-β1 in the pathogenesis of sepsis. RESULTS TGF-β1 level was gradually increased in the lung during the progress of sepsis, and platelets are the major source of the elevated TGF-β1 level in the lung after sepsis. Deficiency of platelet-derived TGF-β1 prolonged the survival of sepsis mice, inhibited the drop of platelet number and bacterial growth, impaired the thrombus formation in the lung and liver, and improved liver function. In addition, platelet TGF-β1 deficiency also decreased the recruitment of neutrophils and monocytes to the lung and impaired neutrophil extracellular trap formation. However, the adoptive transfer of normal platelets to platelet-specific TGF-β1 knockout mice significantly reduced the number of circulating platelets, increased thrombosis in the lung and liver, and promoted the neutrophil extracellular trap formation. CONCLUSIONS Deficiency of platelet-derived TGF-β1 inhibits septic thrombosis and prolongs survival time, indicating that it might be a novel therapeutic target for the treatment of sepsis.
Collapse
Affiliation(s)
- Yingying Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Huimin Jiang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Xinyi Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Hui Zhu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Yue Dai
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Jie Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Yueyue Sun
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Xiang Chu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Wen Ju
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Mengdi Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Lingyu Zeng
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Jianlin Qiao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, China. Blood Diseases Institute, Xuzhou Medical University, China. Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| |
Collapse
|
7
|
Fu KY, Chen F, Jin L, Li GQ. Activin β Is Critical for Larval-Pupal Transition in the 28 Spotted Lady Beetle Henosepilachna vigintioctopunctata. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2025; 118:e70025. [PMID: 39835508 DOI: 10.1002/arch.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/25/2024] [Accepted: 12/30/2024] [Indexed: 01/22/2025]
Abstract
The activin cascade is activated when a pair of extracellular ligand (Myoglianin, Myo; Activin β, Actβ; Dawdle, Daw) binds to two pairs of transforming growth factor β (TGF) serine-threonine receptor kinases, TGF-β type I (Baboon, Babo) and II receptors. However, the roles of activin way have not well been explored in non-Drosophilid insects. In the present paper, we compared the functions of Activin β (Actβ) ligand and receptor isoform BaboB in post-embryonic development in a defoliating ladybird Henosepilachna vigintioctopunctata. RNA interference (RNAi) for Hvactβ but not Hvbabob upregulated juvenile hormone signal, reduced ecdysone pathway and impaired larval-pupal transformation. The arresting Hvactβ RNAi larvae formed pupa-specific black markings below the larval exuviae. Thus, the impairment of metamorphosis may be caused by failing to complete ecdysis behavior due to nonfunctional muscles. Consistently, larval body sizes were smaller and adult appendages were shorter in the Hvactβ RNAi larvae, in contrast to those in the Hvbabob depleted beetles. Conversely, knockdown of Hvbabob but not Hvactβ changed the pigmentation of adult elytra. Our results suggest that Actβ exerts regulative roles in JH production, ecdysteroidogenesis and organ remodeling, thus contributing to modulate the larva-pupa-adult transformation, through a BaboB independent way in H. vigintioctopunctata.
Collapse
Affiliation(s)
- Kai-Yun Fu
- Xinjiang Key Laboratory of Agricultural Biosafety, Institute of Plant Protection, Xinjiang Academy of Agricultural Sciences, Urumqi, Xinjiang, China
- Key Laboratory of Intergraded Management of Harmful Crop Vermin of China North-western Oasis, Ministry of Agriculture, Urumqi, China
| | - Feng Chen
- State Key Laboratory of Agricultural and Forestry Biosecurity, Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Lin Jin
- State Key Laboratory of Agricultural and Forestry Biosecurity, Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Guo-Qing Li
- State Key Laboratory of Agricultural and Forestry Biosecurity, Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
8
|
Mgwenya TN, Abrahamse H, Houreld NN. Photobiomodulation studies on diabetic wound healing: An insight into the inflammatory pathway in diabetic wound healing. Wound Repair Regen 2025; 33:e13239. [PMID: 39610015 PMCID: PMC11628774 DOI: 10.1111/wrr.13239] [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: 04/03/2024] [Revised: 08/06/2024] [Accepted: 09/02/2024] [Indexed: 11/30/2024]
Abstract
Diabetes mellitus remains a global challenge to public health as it results in non-healing chronic ulcers of the lower limb. These wounds are challenging to heal, and despite the different treatments available to improve healing, there is still a high rate of failure and relapse, often necessitating amputation. Chronic diabetic ulcers do not follow an orderly progression through the wound healing process and are associated with a persistent inflammatory state characterised by the accumulation of pro-inflammatory macrophages, cytokines and proteases. Photobiomodulation has been successfully utilised in diabetic wound healing and involves illuminating wounds at specific wavelengths using predominantly light-emitting diodes or lasers. Photobiomodulation induces wound healing through diminishing inflammation and oxidative stress, among others. Research into the application of photobiomodulation for wound healing is current and ongoing and has drawn the attention of many researchers in the healthcare sector. This review focuses on the inflammatory pathway in diabetic wound healing and the influence photobiomodulation has on this pathway using different wavelengths.
Collapse
Affiliation(s)
- Tintswalo N. Mgwenya
- Laser Research Centre, Faculty of Health SciencesUniversity of JohannesburgJohannesburgGautengSouth Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health SciencesUniversity of JohannesburgJohannesburgGautengSouth Africa
| | - Nicolette N. Houreld
- Laser Research Centre, Faculty of Health SciencesUniversity of JohannesburgJohannesburgGautengSouth Africa
| |
Collapse
|
9
|
Zheng Y, Zhang F, Nie H, Li X, Xun J, Fu J, Wu L. Small molecule valproic acid enhances ventral patterning of human neural tube organoids by regulating Wnt and Shh signalling. Cell Prolif 2025; 58:e13737. [PMID: 39164046 PMCID: PMC11693559 DOI: 10.1111/cpr.13737] [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: 05/16/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/22/2024] Open
Abstract
Valproic acid (VPA), a clinically approved small molecule, has been reported to activate Wnt signalling that is critical for dorsal-ventral (DV) patterning of neural tube. However, little is known about the impact of VPA on DV patterning process. Here, we show that even though VPA has a negative impact on the early formation of human neural tube organoids (hNTOs), it significantly enhances the efficiency of ventrally patterned hNTOs, when VPA is added during the entire differentiation process. RNA sequencing and RT-qPCR analysis demonstrates VPA activates endogenous Wnt signalling in hNTOs. Surprisingly, transcriptome analysis also identifies upregulation of genes for degradation of GLI2 and GLI3 proteins, whose truncated fragment are transcriptional repressors of Shh signalling. The Western-blot analysis confirms the increase of GLI3R proteins after VPA treatment. Thus, VPA might enhance ventral patterning of hNTOs through both activating Wnt, which can antagonise Shh signalling by inducing GLI3 expression, and/or inhibiting Shh signalling by inducing GLI protein degradation. We further obtain results to show that VPA still increases patterning efficiency of hNTOs with a weak influence on their early formation when the initiation time of VPA is delayed and its duration is reduced. Taken together, this study demonstrates that VPA enhances the generation of more reproducible hNTOs with ventral patterning, opening the avenues for the applications of hNTOs in developmental biology and regenerative medicine.
Collapse
Affiliation(s)
- Yuanyuan Zheng
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institute of Physical Science and Information TechnologyAnhui UniversityHefeiChina
| | - Fangrong Zhang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institute of Physical Science and Information TechnologyAnhui UniversityHefeiChina
| | - Haifeng Nie
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institute of Physical Science and Information TechnologyAnhui UniversityHefeiChina
| | - Xinyu Li
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institute of Physical Science and Information TechnologyAnhui UniversityHefeiChina
| | - Jiali Xun
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institute of Physical Science and Information TechnologyAnhui UniversityHefeiChina
| | - Jianping Fu
- Department of Mechanical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of Cell & Developmental BiologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Lijun Wu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institute of Physical Science and Information TechnologyAnhui UniversityHefeiChina
| |
Collapse
|
10
|
Fakhari S, Campolina‐Silva G, Asayesh F, Girardet L, Scott‐Boyer M, Droit A, Soulet D, Greener J, Belleannée C. Shear stress effects on epididymal epithelial cell via primary cilia mechanosensory signaling. J Cell Physiol 2025; 240:e31475. [PMID: 39508588 PMCID: PMC11733861 DOI: 10.1002/jcp.31475] [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: 07/03/2024] [Revised: 09/24/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024]
Abstract
Shear stress, resulting from fluid flow, is a fundamental mechanical stimulus affecting various cellular functions. The epididymis, essential for sperm maturation, offers a compelling model to study the effects of shear stress on cellular behavior. This organ undergoes extensive proliferation and differentiation until puberty, achieving full functionality as spermatozoa commence their post-testicular maturation. Although the mechanical tension exerted by testicular fluid is hypothesized to drive epithelial proliferation and differentiation, the precise mechanisms remain unclear. Here we assessed whether the responsiveness of the epididymal cells to shear stress depends on functional primary cilia by combining microfluidic strategies on immortalized epididymal cells, calcium signaling assays, and high-throughput gene expression analysis. We identified 97 genes overexpressed in response to shear stress, including early growth response (Egr) 2/3, cellular communication network factor (Ccn) 1/2, and Fos proto-oncogene (Fos). While shear stress triggered a rapid increase of intracellular Ca2+, this response was abrogated following the impairment of primary ciliogenesis through pharmacological and siRNA approaches. Overall, our findings provide valuable insights into how mechanical forces influence the development of the male reproductive system, a requisite to sperm maturation.
Collapse
Affiliation(s)
- Sepideh Fakhari
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
- Department of ChemistryFaculty of Science and EngineeringQuébec CityQuebecCanada
| | - Gabriel Campolina‐Silva
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
| | - Farnaz Asayesh
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
| | - Laura Girardet
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
| | - Marie‐Pier Scott‐Boyer
- Proteomics Platform, Québec Genomic Center, Université Laval, CHU de Québec Research Center (CHUL)Québec CityQuebecCanada
| | - Arnaud Droit
- Proteomics Platform, Québec Genomic Center, Université Laval, CHU de Québec Research Center (CHUL)Québec CityQuebecCanada
| | - Denis Soulet
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
- Faculté de pharmacieUniversité LavalQuébec CityQuebecCanada
| | - Jesse Greener
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
- Department of ChemistryFaculty of Science and EngineeringQuébec CityQuebecCanada
| | - Clémence Belleannée
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
| |
Collapse
|
11
|
Jahan N, Islam S, Sivasundaram K, Ota A, Naito M, Kuroda J, Watanabe H. Role of versican in extracellular matrix formation: analysis in 3D culture. Am J Physiol Cell Physiol 2025; 328:C245-C257. [PMID: 39656505 DOI: 10.1152/ajpcell.00495.2024] [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: 07/15/2024] [Revised: 10/28/2024] [Accepted: 11/20/2024] [Indexed: 01/01/2025]
Abstract
Three-dimensional (3-D) cell culture creates an environment that allows cells to grow and interact with the surrounding extracellular framework. Versican plays a pivotal role in forming the provisional matrix, but it is still unclear how this proteoglycan affects the formation of the extracellular matrix. Here, we established a 3-D culture system using fibrin gel, which enables a long-term culture up to a month. With this system, we characterized fibroblasts obtained from the newborn knock-in homozygotes, termed R/R, expressing a disintegrin and metalloproteinase with thrombospondin motif (ADAMTS)-resistant versican and wild-type mice. R/R fibroblasts showed higher levels of versican deposition than wild-type, demonstrating that the initial ADAMTS-cleavage site is involved in versican turnover. These fibroblasts exhibited faster proliferation and myofibroblastic differentiation, concomitant with higher levels of transforming growth factor β-signaling. R/R fibroblast culture had higher deposition levels of fibronectin, type I and V collagens, and fibrillin-1, especially at the late stages of culture. These results suggest that versican expressed by dermal fibroblasts facilitates the extracellular matrix formation, at least by affecting fibroblast behavior.NEW & NOTEWORTHY We established a 3-D-culture system useful for analyzing fibroblast behavior and matrix formation. The initial cleavage site by ADAMTSs in versican core protein is mainly involved in versican turnover. Accumulating versican facilitates fibroblast proliferation and myofibroblastic differentiation in an autocrine or paracrine manner. Accumulating versican promotes the deposition of fibronectin and collagens.
Collapse
Affiliation(s)
- Nushrat Jahan
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| | - Shamima Islam
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| | - Karnan Sivasundaram
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Japan
| | - Akinobu Ota
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Japan
| | - Munekazu Naito
- Department of Anatomy, School of Medicine, Aichi Medical University, Nagakute, Japan
| | - Junpei Kuroda
- Laboratory of Pattern Formation, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
12
|
Saraswati W, Yahya AN, Yonas Y, Cindananti G, Rahardia N, Ramadani RR, Ambarwati VL, Kusumawardhani DP, Sutela IGMY, Putri LK, Baskoro BD, Pratama PK, Setiady DR. Calcium Carbonate from Anadara granosa Shells Stimulates FGF2, TGF-β1, and Collagen Type 1 Expression in Rat Dental Pulp. Eur J Dent 2024. [PMID: 39750522 DOI: 10.1055/s-0044-1793842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVES Calcium carbonate (CaCO3), a major inorganic component in bones and teeth, offers potential protection against demineralization. This study investigates the effect of CaCO3 from Anadara granosa shells on the expression of fibroblast growth factor 2 (FGF2), transforming growth factor-β1 (TGF-β1), and collagen type 1 in the rat dental pulp. MATERIALS AND METHODS The first maxillary molars of Rattus norvegicus were perforated and subsequently pulp capped with CaCO3 extracted from A. granosa shells. The cavities were then filled with glass ionomer cement, while the control group received calcium hydroxide (Ca(OH)2). Teeth were extracted after 7 and 14 days of treatment, and the expression of FGF2, TGF-β1, and collagen type 1 in the dental pulp was analyzed using immunohistochemistry staining. RESULTS The group treated with CaCO3 from A. granosa shells exhibited significantly higher expression of FGF2, TGF-β1, and collagen type 1 in the dental pulp at both 7 and 14 days compared with the group treated with Ca(OH)2 (p < 0.01). CONCLUSION The application of CaCO3 derived from A. granosa shells enhances the proliferative phase in the dental pulp after pulp perforation and perhaps promotes reparative dentine formation.
Collapse
Affiliation(s)
- Widya Saraswati
- Department of Conservative Dentistry, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Azlin Noor Yahya
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Malaya, Malaysia
| | - Yovita Yonas
- Postgraduate Program of Conservative Dentistry Specialist, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ganiezha Cindananti
- Postgraduate Program of Conservative Dentistry Specialist, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Nabiela Rahardia
- Postgraduate Program of Conservative Dentistry Specialist, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Rania Rizka Ramadani
- Postgraduate Program of Conservative Dentistry Specialist, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Venny Lusanda Ambarwati
- Postgraduate Program of Conservative Dentistry Specialist, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | | | | | - Larasati Kianti Putri
- Postgraduate Program of Conservative Dentistry Specialist, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Brian Dwi Baskoro
- Postgraduate Program of Conservative Dentistry Specialist, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Putu Krisnanda Pratama
- Postgraduate Program of Conservative Dentistry Specialist, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Dawailatur Rahman Setiady
- Postgraduate Program of Conservative Dentistry Specialist, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
13
|
Miyajima C, Nagasaka M, Aoki H, Toriuchi K, Yamanaka S, Hashiguchi S, Morishita D, Aoyama M, Hayashi H, Inoue Y. The Hippo Signaling Pathway Manipulates Cellular Senescence. Cells 2024; 14:13. [PMID: 39791714 PMCID: PMC11719916 DOI: 10.3390/cells14010013] [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: 10/28/2024] [Revised: 12/21/2024] [Accepted: 12/24/2024] [Indexed: 01/12/2025] Open
Abstract
The Hippo pathway, a kinase cascade, coordinates with many intracellular signals and mediates the regulation of the activities of various downstream transcription factors and their coactivators to maintain homeostasis. Therefore, the aberrant activation of the Hippo pathway and its associated molecules imposes significant stress on tissues and cells, leading to cancer, immune disorders, and a number of diseases. Cellular senescence, the mechanism by which cells counteract stress, prevents cells from unnecessary damage and leads to sustained cell cycle arrest. It acts as a powerful defense mechanism against normal organ development and aging-related diseases. On the other hand, the accumulation of senescent cells without their proper removal contributes to the development or worsening of cancer and age-related diseases. A correlation was recently reported between the Hippo pathway and cellular senescence, which preserves tissue homeostasis. This review is the first to describe the close relationship between aging and the Hippo pathway, and provides insights into the mechanisms of aging and the development of age-related diseases. In addition, it describes advanced findings that may lead to the development of tissue regeneration therapies and drugs targeting rejuvenation.
Collapse
Affiliation(s)
- Chiharu Miyajima
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
| | - Mai Nagasaka
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
- Department of Experimental Chemotherapy, Cancer Chemotherapy Center of JFCR, Tokyo 135-8550, Japan
| | - Hiromasa Aoki
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (H.A.); (K.T.); (M.A.)
| | - Kohki Toriuchi
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (H.A.); (K.T.); (M.A.)
| | - Shogo Yamanaka
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
| | - Sakura Hashiguchi
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
| | - Daisuke Morishita
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
| | - Mineyoshi Aoyama
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (H.A.); (K.T.); (M.A.)
| | - Hidetoshi Hayashi
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
| | - Yasumichi Inoue
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; (M.N.); (S.Y.); (S.H.); (D.M.); (H.H.)
| |
Collapse
|
14
|
Hosseini Farzad S, Lashkarboloki M, Mowla SJ, Soltani BM. LncRNA DANCR-V1 is a novel regulator of Wnt/β-catenin and TGF-β1/SMAD signaling pathways in colorectal cancer: an in vitro and in silico study. Mol Biol Rep 2024; 52:36. [PMID: 39643825 DOI: 10.1007/s11033-024-10128-0] [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: 08/26/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND DANCR is an oncogenic lncRNA associated with advanced colorectal cancer, one of the most common malignancies worldwide. This lncRNA has a new variant, DANCR-V1, whose function is not yet understood. In this study, we aimed to evaluate the expression pattern of DANCR-V1 and its regulatory mechanism in colorectal cancer. METHOD AND RESULT Bioinformatics analysis and RT-qPCR showed that DANCR-V1 expression was higher in colorectal cancer tissues than in normal pairs obtained from microarray data and 20 samples, respectively. LncRNA subcellular localization and hsa-miR-222 binding sites were predicted using bioinformatics tools. Dual luciferase assays confirmed that miR-222-mediated downregulation of DANCR-V1 through its targeting, and RT-qPCR showed that overexpression of miR-222 decreased the level of DANCR-V1. Functionally, Wnt/β-catenin and TGF-β1/SMAD-related genes changed under DANCR-V1 overexpression in the SW480 cell line, while their expression was reversed following miR-222 overexpression. Finally, at the cellular level, overexpression of DANCR-V1 elevated the proliferation and migration rates of SW480 cells, as determined using flow cytometry, western blotting and scratch assays. CONCLUSION Our data suggest that DANCR-V1 is a novel transcript variant that has crucial crosstalk with miR-222 via negative feedback and plays a critical role in colorectal cancer progression through Wnt/β-catenin and TGF-β1/SMAD signaling modulation.
Collapse
Affiliation(s)
- Sana Hosseini Farzad
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mina Lashkarboloki
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Javad Mowla
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahram M Soltani
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
15
|
Wu L, Wang J, Chai L, Chen J, Jin X. Roles of deubiquitinases in urologic cancers (Review). Oncol Lett 2024; 28:609. [PMID: 39525605 PMCID: PMC11544529 DOI: 10.3892/ol.2024.14743] [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: 07/23/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Human health is endangered by the occurrence and progression of urological cancers, including renal cell carcinoma, prostate cancer and bladder cancer, which are usually associated with the activation of oncogenic factors and inhibition of cancer suppressors. The primary mechanism for protein breakdown in cells is the ubiquitin-proteasome system, whilst deubiquitinases contribute to the reversal of this process. However, both are important for protein homeostasis. Deubiquitination may also be involved in the control of the cell cycle, proliferation and apoptosis, and dysregulated deubiquitination is associated with the malignant transformation, invasion and metastasis of urologic malignancies. Therefore, a comprehensive summary of the mechanisms underlying deubiquitination in urological cancers may provide novel strategies and insights for diagnosis and treatment. The present review aimed to methodically clarify the role of deubiquitinating enzymes in urinary system cancers as well as their prospective application prospects for clinical treatment.
Collapse
Affiliation(s)
- Liangpei Wu
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jiahui Wang
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Lin Chai
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jun Chen
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
16
|
Gao S, Wang X, Huang Y, You L. Calreticulin-driven autophagy enhances cell proliferation in laryngeal squamous cell carcinoma. Tissue Cell 2024; 91:102603. [PMID: 39550898 DOI: 10.1016/j.tice.2024.102603] [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: 04/02/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Calreticulin (CALR) is a multifunctional calcium-binding protein. Recent studies have revealed that CALR contributes to tumor development and promotes cancer cell proliferation. However, how CALR affects the development of laryngeal squamous cell carcinoma (LSCC) remains mysterious. Thus, this study aimed to explore the effect of CALR on LSCC development and uncover its underlying mechanisms. METHODS CALR expression in LSCC cell lines and tissues was examined by qRT-PCR and western blot analysis and its functional role was detected via in vivo and in vitro assays. Cell proliferation was discriminated with CCK-8 and colony formation assays, while apoptosis was analyzed using flow cytometry. Autophagy levels were measured via LC3 immunofluorescence, and western blot assay was conducted to assess apoptosis- and autophagy-related proteins. Additionally, a mouse xenograft model was employed to determine the impact of CALR knockdown on tumor growth. RESULTS We found that CALR knockdown reduced LSCC cell viability and proliferation while enhancing apoptosis, whereas CALR overexpression showed opposite effects. In vivo experiments verified that CALR knockdown suppressed tumor growth. In addition, elevated CALR expression induced autophagy in LSCC cells, while autophagy inhibitor 3-MA (2.5 mM) reversed the anti-apoptosis effects of CALR overexpression. CONCLUSION Our study identifies CALR as an oncogene in LSCC, where it promotes tumor progression by inducing autophagy and inhibiting apoptosis. Targeting CALR or modulating autophagy may represent novel therapeutic strategies for LSCC.
Collapse
Affiliation(s)
- Shufeng Gao
- Department of ENT & HN Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China.
| | - Xintao Wang
- Department of ENT & HN Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Yun Huang
- Department of ENT & HN Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Longgui You
- Department of ENT & HN Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| |
Collapse
|
17
|
Jang Y, Kang S, Han H, Kang CM, Cho NH, Kim BG. Fibrosis-Encapsulated Tumoroid, A Solid Cancer Assembloid Model for Cancer Research and Drug Screening. Adv Healthc Mater 2024; 13:e2402391. [PMID: 39233539 PMCID: PMC11650424 DOI: 10.1002/adhm.202402391] [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: 06/30/2024] [Revised: 08/12/2024] [Indexed: 09/06/2024]
Abstract
Peritumoral fibrosis is known to promote cancer progression and confer treatment resistance in various solid tumors. Consequently, developing accurate cancer research and drug screening models that replicate the structure and function of a fibrosis-surrounded tumor mass is imperative. Previous studies have shown that self-assembly three-dimensional (3D) co-cultures primarily produce cancer-encapsulated fibrosis or maintain a fibrosis-encapsulated tumor mass for a short period, which is inadequate to replicate the function of fibrosis, particularly as a physical barrier. To address this limitation, a multi-layer spheroid formation method is developed to create a fibrosis-encapsulated tumoroid (FET) structure that maintains structural stability for up to 14 days. FETs exhibited faster tumor growth, higher expression of immunosuppressive cytokines, and equal or greater resistance to anticancer drugs compared to their parental tumoroids. Additionally, FETs serve as a versatile model for traditional cancer research, enabling the study of exosomal miRNA and gene functions, as well as for mechanobiology research when combined with alginate hydrogel. Our findings suggest that the FET represents an advanced model that more accurately mimics solid cancer tissue with peritumoral fibrosis. It may show potential superiority over self-assembly-based 3D co-cultures for cancer research and drug screening, and holds promise for personalized drug selection in cancer treatment.
Collapse
Affiliation(s)
- Yeonsue Jang
- Department of Urological Science InstituteYonsei University College of MedicineSeoul03722Republic of Korea
| | - Suki Kang
- Department of PathologyYonsei University College of MedicineSeoul03722Republic of Korea
| | - Hyunho Han
- Department of Urological Science InstituteYonsei University College of MedicineSeoul03722Republic of Korea
- Division of Hepatobiliary and Pancreatic Surgery, Department of SurgeryYonsei University College of MedicineSeoul03722Republic of Korea
| | - Chang Moo Kang
- Division of Hepatobiliary and Pancreatic Surgery, Department of SurgeryYonsei University College of MedicineSeoul03722Republic of Korea
| | - Nam Hoon Cho
- Department of PathologyYonsei University College of MedicineSeoul03722Republic of Korea
- Brain Korea 21 Plus Project for Medical ScienceYonsei University College of MedicineSeoul03722South Korea
| | - Baek Gil Kim
- Department of PathologyYonsei University College of MedicineSeoul03722Republic of Korea
- Brain Korea 21 Plus Project for Medical ScienceYonsei University College of MedicineSeoul03722South Korea
| |
Collapse
|
18
|
Zheng Y, Li L, Cai W, Li L, Zhang R, Huang W, Cao Y. Unveiling the role of TGF-β signaling pathway in breast cancer prognosis and immunotherapy. Front Oncol 2024; 14:1488137. [PMID: 39664194 PMCID: PMC11631921 DOI: 10.3389/fonc.2024.1488137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction The TGF-β signaling pathway (TSP) is pivotal in tumor progression. Nonetheless, the connection between genes associated with the TSP and the clinical outcomes of breast cancer, as well as their impact on the tumor microenvironment and immunotherapeutic responses, remains elusive. Methods Breast cancer transcriptomic and single-cell sequencing data were obtained from the The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. We identified 54 genes associated with the TSP from the Molecular Signatures Database (MSigDB) and analyzed both data types to evaluate TSP activity. Using weighted gene co-expression network analysis (WGCNA), we identified modules linked to TSP activity. To assess patient risk, we used 101 machine learning algorithms to develop an optimal TGF-β pathway-related prognostic signature (TSPRS). We then examined immune activity and response to immune checkpoint inhibitors and chemotherapy in these groups. Finally, we validated ZMAT3 expression levels clinically and confirmed its relevance in breast cancer using CCK-8 and migration assays. Results At the single-cell level, TSP activity was most notable in endothelial cells, with higher activity in normal tissues compared to tumors. TSPRS was developed. This signature's accuracy was confirmed through internal and external validations. A nomogram incorporating the TSPRS was created to improve prediction accuracy. Further studies showed that breast cancer patients categorized as low-risk by the TSPRS had higher immune phenotype scores and more immune cell infiltration, leading to better prognosis and enhanced immunotherapy response. Additionally, a strong link was found between the TSPRS risk score and the effectiveness of anti-tumor agents. Silencing the ZMAT3 gene in the TSPRS significantly reduced the proliferation and invasiveness of breast cancer cells. Discussion Our study developed a TSPRS, which emerges as a potent predictive instrument for the prognosis of breast cancer, offering novel perspectives on the immunotherapeutic approach to the disease.
Collapse
Affiliation(s)
- Yifan Zheng
- Department of General Surgery I, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Li Li
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenqian Cai
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lin Li
- Department of General Surgery I, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Rongxin Zhang
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Institute of Basic Medical Sciences and Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenbin Huang
- Department of General Surgery I, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yulun Cao
- Department of General Surgery I, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Pan Z, Yao Q, Kong W, Ma X, Tian L, Zhao Y, Zhu S, Chen S, Sun M, Liu J, Jiang S, Ma J, Liu Q, Peng X, Li X, Hong Z, Hong Y, Wang X, Liu J, Zhang J, Zhang W, Sun B, Pahlavan S, Xia Y, Shen W, Liu Y, Jiang W, Xie Z, Kong W, Wang X, Wang K. Generation of iPSC-derived human venous endothelial cells for the modeling of vascular malformations and drug discovery. Cell Stem Cell 2024:S1934-5909(24)00377-1. [PMID: 39579761 DOI: 10.1016/j.stem.2024.10.015] [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: 05/13/2024] [Revised: 09/04/2024] [Accepted: 10/28/2024] [Indexed: 11/25/2024]
Abstract
Venous malformations (VMs) represent prevalent vascular anomalies typically attributed to non-inherited somatic mutations within venous endothelial cells (VECs). The lack of robust disease models for VMs impedes drug discovery. Here, we devise a robust protocol for the generation of human induced VECs (iVECs) through manipulation of cell-cycle dynamics via the retinoic signaling pathway. We introduce an L914F mutation into the TIE2 gene locus of induced pluripotent stem cells (iPSCs) and show that the mutated iVECs form dilated blood vessels after transplantation into mice, thereby recapitulating the phenotypic characteristics observed in VMs. Moreover, utilizing a deep neural network and a high-throughput digital RNA with perturbation of genes sequencing (DRUG-seq) approach, we perform drug screening and demonstrate that bosutinib effectively rescues the disease phenotype in vitro and in vivo. In summary, by leveraging genome editing and stem cell technology, we generate VM models that enable the development of additional therapeutics.
Collapse
Affiliation(s)
- Zihang Pan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Qiyang Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Weijing Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xiaojing Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Liangliang Tian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Yun Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Shuntian Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Sheng Chen
- Department of Burns and Plastic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mengze Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Jiao Liu
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing 100191, China
| | - Simin Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Jianxun Ma
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Qijia Liu
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Xiaohong Peng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xiaoxia Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Zixuan Hong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Yi Hong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xue Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Jiarui Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Jingjing Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Wei Zhang
- TianXinFu (Beijing) Medical Appliance Co., Ltd., Beijing 102200, China
| | - Bingbing Sun
- TianXinFu (Beijing) Medical Appliance Co., Ltd., Beijing 102200, China
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Youchen Xia
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Weimin Shen
- Department of Burns and Plastic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuyong Liu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Wenjian Jiang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Zhengwei Xie
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing 100191, China.
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China.
| | - Xi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China; State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China.
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China.
| |
Collapse
|
20
|
Heath J, Mirabelli C, Annis MG, Sabourin V, Hebert S, Findlay S, Kim H, Witcher M, Kleinman CL, Siegel PM, Orthwein A, Ursini-Siegel J. The Neurodevelopmental Protein POGZ Suppresses Metastasis in Triple-Negative Breast Cancer by Attenuating TGFβ Signaling. Cancer Res 2024; 84:3743-3760. [PMID: 39137399 DOI: 10.1158/0008-5472.can-23-3887] [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: 12/07/2023] [Revised: 06/03/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024]
Abstract
The pogo transposable element-derived zinc finger protein, POGZ, is notably associated with neurodevelopmental disorders through its role in gene transcription. Many proteins involved in neurological development are often dysregulated in cancer, suggesting a potential role for POGZ in tumor biology. Here, we provided experimental evidence that POGZ influences the growth and metastatic spread of triple-negative breast cancers (TNBC). In well-characterized models of TNBC, POGZ exerted a dual role, both as a tumor promoter and metastasis suppressor. Mechanistically, loss of POGZ potentiated TGFβ pathway activation to exert cytostatic effects while simultaneously increasing the mesenchymal and migratory properties of breast tumors. Although POGZ levels are elevated in human breast cancers, the most aggressive forms of TNBC tumors, including those with increased mesenchymal and metastatic properties, exhibit dampened POGZ levels, and low POGZ expression was associated with inferior clinical outcomes in these tumor types. Taken together, these data suggest that POGZ is a critical suppressor of the early stages of the metastatic cascade. Significance: The POGZ neurodevelopmental protein plays dual functions in triple-negative breast cancers as a tumor promoter and metastasis suppressor, inhibiting TGFβ-regulated EMT to limit breast cancer metastatic progression.
Collapse
Affiliation(s)
- John Heath
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Caitlynn Mirabelli
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Matthew G Annis
- Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Valerie Sabourin
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Steven Hebert
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Steven Findlay
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - HaEun Kim
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Michael Witcher
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Claudia L Kleinman
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Peter M Siegel
- Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Alexandre Orthwein
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Josie Ursini-Siegel
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
21
|
Jardanowska-Kotuniak M, Dramiński M, Własnowolski M, Łapiński M, Sengupta K, Agarwal A, Filip A, Ghosh N, Pancaldi V, Grynberg M, Saha I, Plewczynski D, Dąbrowski MJ. Unveiling epigenetic regulatory elements associated with breast cancer development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623187. [PMID: 39605637 PMCID: PMC11601335 DOI: 10.1101/2024.11.12.623187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Breast cancer is the most common cancer in women and the 2nd most common cancer worldwide, yearly impacting over 2 million females and causing 650 thousand deaths. It has been widely studied, but its epigenetic variation is not entirely unveiled. We aimed to identify epigenetic mechanisms impacting the expression of breast cancer related genes to detect new potential biomarkers and therapeutic targets. We considered The Cancer Genome Atlas database with over 800 samples and several omics datasets such as mRNA, miRNA, DNA methylation, which we used to select 2701 features that were statistically significant to differ between cancer and control samples using the Monte Carlo Feature Selection and Interdependency Discovery algorithm, from an initial total of 417,486. Their biological impact on cancerogenesis was confirmed using: statistical analysis, natural language processing, linear and machine learning models as well as: transcription factors identification, drugs and 3D chromatin structure analyses. Classification of cancer vs control samples on the selected features returned high classification weighted Accuracy from 0.91 to 0.98 depending on feature-type: mRNA, miRNA, DNA methylation, and classification algorithm. In general, cancer samples showed lower expression of differentially expressed genes and increased β-values of differentially methylated sites. We identified mRNAs whose expression is well explained by miRNA expression and differentially methylated sites β-values. We recognized differentially methylated sites possibly affecting NRF1 and MXI1 transcription factors binding, causing a disturbance in NKAPL and PITX1 expression, respectively. Our 3D models showed more loosely packed chromatin in cancer. This study successfully points out numerous possible regulatory dependencies.
Collapse
Affiliation(s)
- Marta Jardanowska-Kotuniak
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
- Institute of Biochemistry and Biophysics of the Polish Academy of Sciences, Warsaw, Poland
| | - Michał Dramiński
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Michał Własnowolski
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | - Marcin Łapiński
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Kaustav Sengupta
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | - Abhishek Agarwal
- Laboratory of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Adam Filip
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Nimisha Ghosh
- Department of Computer Science and Information Technology, Institute of Technical Education and Research, Siksha O Anusandhan University, Bhubaneswar, Odisha, 751030, India
| | - Vera Pancaldi
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Marcin Grynberg
- Institute of Biochemistry and Biophysics of the Polish Academy of Sciences, Warsaw, Poland
| | - Indrajit Saha
- Department of Computer Science and Engineering, National Institute of Technical Teachers’ Training and Research, Kolkata 700106, India
| | - Dariusz Plewczynski
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
- Laboratory of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Michał J. Dąbrowski
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
22
|
Islam P, Abosalha A, Schaly S, Boyajian JL, Santos M, Makhlouf S, Renesteen E, Kassab A, Shum-Tim C, Shum-Tim D, Prakash S. Baculovirus Expressing Tumor Growth Factor-β1 (TGFβ1) Nanoshuttle Augments Therapeutic Effects for Vascular Wound Healing: Design and In Vitro Analysis. ACS Pharmacol Transl Sci 2024; 7:3419-3428. [PMID: 39539270 PMCID: PMC11555499 DOI: 10.1021/acsptsci.4c00509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/20/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
One of the major challenges in vascular tissue regeneration is effective wound healing that can be resolved by an innovative targeted nanoshuttle that delivers growth factors to blood vessels. This study investigates the production and efficacy of transforming growth factor-β1 (TGFβ1) gene delivery using poly(lactic-co-glycolic acid) (PLGA) baculovirus (BV) nanoshuttles (NSs). They exhibited an encapsulation efficiency of 86.23% ± 0.65% and a negative zeta potential of -29.57 ± 1.27 mV. In vitro studies in human umbilical vein endothelial cells (HUVECs) revealed that a 12 h incubation period optimized virus transduction. The safety and superior intracellular uptake of NSs and BVs in HUVECs were observed. The NSs carrying 100 and 400 MOI exhibited the highest cell proliferation rates in HUVECs. These sustained-release NSs significantly improved vascular cell migration and wound closure compared to free TGFβ1 carrying BV and can be a groundbreaking find in regenerative medicine, cardiovascular diseases, and chronic ulcer conditions.
Collapse
Affiliation(s)
- Paromita Islam
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Ahmed Abosalha
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
- Pharmaceutical
Technology Department, Faculty of Pharmacy, Tanta University, Tanta
Al-Geish St., the Medical Campus, Tanta 31527, Egypt
| | - Sabrina Schaly
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jacqueline L. Boyajian
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Madison Santos
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Stephanie Makhlouf
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Editha Renesteen
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Amal Kassab
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Cedrique Shum-Tim
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
- Mila−Quebec
AI Institute, McGill University, 6666 Saint-Urbain Street, Montreal, Quebec H2S 3H1, Canada
| | - Dominique Shum-Tim
- Division
of Cardiac Surgery, Royal Victoria Hospital, McGill University Health
Centre, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3G 2M1, Canada
| | - Satya Prakash
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
23
|
Al-Regaiey K. Crosstalk between adipogenesis and aging: role of polyphenols in combating adipogenic-associated aging. Immun Ageing 2024; 21:76. [PMID: 39511615 PMCID: PMC11542427 DOI: 10.1186/s12979-024-00481-w] [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: 08/17/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024]
Abstract
In the last forty years, the number of people over 60 years of age has increased significantly owing to better nutrition and lower rates of infectious diseases in developing countries. Aging significantly impacts adipose tissue, which plays crucial role in hormone regulation and energy storage. This can lead to imbalances in glucose, and overall energy homeostasis within the body. Aging is irreversible phenomena and potentially causing lipid infiltration in other organs, leading to systemic inflammation, metabolic disorders. This review investigates various pathways contributing to aging-related defects in adipogenesis, such as changes in adipose tissue function and distribution. Polyphenols, a diverse group of natural compounds, can mitigate aging effects via free radicals, oxidative stress, inflammation, senescence, and age-related diseases. Polyphenols like resveratrol, quercetin and EGCG exhibit distinct mechanisms and regulate crucial pathways, such as the TGF-β, AMPK, Wnt, PPAR-γ, and C/EBP transcription factors, and influence epigenetic modifications, such as DNA methylation and histone modification. This review highlights the critical importance of understanding the intricate relationship between aging and adipogenesis for optimizing well-being with increasing age. These findings highlight the therapeutic potential of polyphenols like quercetin and resveratrol in enhancing adipose tissue function and promoting healthy aging.
Collapse
Affiliation(s)
- Khalid Al-Regaiey
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
24
|
Papp FR, Katko M, Csiki R, Galgoczi E, Molnar Z, Erdei A, Bodor M, Steiber Z, Ujhelyi B, Nagy EV. Characteristics of Hyaluronan Metabolism During Myofibroblast Differentiation in Orbital Fibroblasts. Invest Ophthalmol Vis Sci 2024; 65:13. [PMID: 39504052 PMCID: PMC11549924 DOI: 10.1167/iovs.65.13.13] [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: 07/30/2024] [Accepted: 10/12/2024] [Indexed: 11/11/2024] Open
Abstract
Purpose To study the impact of myofibroblast differentiation (MD) on hyaluronan (HA) turnover in orbital fibroblasts (OFs) focusing on the expression of its key enzymes and their potential implications in the pathogenesis of thyroid eye disease (TED). Methods Primary cultures of OFs were established from tissue samples (TED OFs, n = 4; non-TED OFs, n = 5). MD was induced by TGF-β1 (5 ng/mL). Measurements were performed after 24- and 72-hour treatments. The proliferation rate was determined by 5-bromo-2'-deoxyuridine (BrdU) incorporation. HA level and size were measured using an aggrecan-based ELISA-like method and agarose gel electrophoresis, respectively. mRNA expressions of myofibroblast markers and enzymes with a role in HA metabolism were determined using real-time PCR. Results Upregulation of type I collagen alpha1 chain, alpha-smooth muscle actin, and fibronectin indicated that OFs underwent MD after stimulation by TGF-β. After 72 hours, proliferation of untreated cultures declined, but it remained higher in myofibroblasts. Pericellular HA content, but not HA in the supernatant of myofibroblasts, increased compared to untreated cells. TGF-β was a potent stimulator of hyaluronan synthase 1 (HAS1) expression. The expression of hyaluronidase-1 and cell migration-inducing protein (CEMIP) diminished following MD, whereas the expression of transmembrane protein 2, the regulator of HA catabolism through CEMIP, was elevated. The size distribution of HA shifted toward a high-molecular-weight form following treatment with TGF-β. Conclusions OFs undergoing MD are characterized by decreased HA turnover as a consequence of the inhibition of hyaluronidases and HAS1 induction. Our results suggest that hyaluronidases could be potential targets in the treatment of TED.
Collapse
Affiliation(s)
- Fruzsina R. Papp
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Health Sciences, University of Debrecen, Debrecen, Hungary
| | - Monika Katko
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Robert Csiki
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Health Sciences, University of Debrecen, Debrecen, Hungary
| | - Erika Galgoczi
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsanett Molnar
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Annamaria Erdei
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklos Bodor
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zita Steiber
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Bernadett Ujhelyi
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Endre V. Nagy
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
25
|
Xia Y, Wang H, Shao M, Liu X, Sun F. MAP3K19 Promotes the Progression of Tuberculosis-Induced Pulmonary Fibrosis Through Activation of the TGF-β/Smad2 Signaling Pathway. Mol Biotechnol 2024; 66:3300-3310. [PMID: 37906388 DOI: 10.1007/s12033-023-00941-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/09/2023] [Indexed: 11/02/2023]
Abstract
Tuberculosis-induced pulmonary fibrosis (PF) is a chronic, irreversible interstitial lung disease, which severely affects lung ventilation and air exchange, leading to respiratory distress, impaired lung function, and ultimately death. As previously reported, epithelial-mesenchymal transition (EMT) and fibrosis in type II alveolar epithelial cells (AEC II) are two critical processes that contributes to the initiation and progression of tuberculosis-related PF, but the underlying pathological mechanisms remain unclear. In this study, through performing Real-Time quantitative PCR (RT-qPCR), Western blot, immunohistochemistry, and immunofluorescence staining assay, we confirmed that the expression levels of EMT and fibrosis-related biomarkers were significantly increased in lung tissues with tuberculosis-associated PF in vivo and Mycobacterium bovis Bacillus Calmette-Guérin (BCG) strain-infected AEC II cells in vitro. Besides, we noticed that the mitogen-activated protein kinase 19 (MAP3K19) was aberrantly overexpressed in PF models, and silencing of MAP3K19 significantly reduced the expression levels of fibronectin, collagen type I, and alpha-smooth muscle actin to decrease fibrosis, and upregulated E-cadherin and downregulated vimentin to suppress EMT in BCG-treated AEC II cells. Then, we uncovered the underlying mechanisms and found that BCG synergized with MAP3K19 to activate the pro-inflammatory transforming growth factor-beta (TGF-β)/Smad2 signal pathway in AEC II cells, and BCG-induced EMT process and fibrosis in AEC II cells were all abrogated by co-treating cells with TGF-β/Smad2 signal pathway inhibitor LY2109761. In summary, our results uncovered the underlying mechanisms by which the MAP3K19/TGF-β/Smad2 signaling pathway regulated EMT and fibrotic phenotypes of AEC II cells to facilitate the development of tuberculosis-associated PF, and these findings will provide new ideas and biomarkers to ameliorate tuberculosis-induced PF in clinic.
Collapse
Affiliation(s)
- Yu Xia
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China.
| | - Haiyue Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China
| | - Meihua Shao
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China
| | - Xuemei Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China
| | - Feng Sun
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China
| |
Collapse
|
26
|
Pak B, Kim M, Han O, Lee HW, Dubrac A, Choi W, Yang JM, Boyé K, Cho H, Citrin KM, Kim I, Eichmann A, Bautch VL, Jin SW. ACVR1/ALK2-p21 signaling axis modulates proliferation of the venous endothelium in the retinal vasculature. Angiogenesis 2024; 27:765-777. [PMID: 38955953 DOI: 10.1007/s10456-024-09936-6] [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: 03/04/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024]
Abstract
The proliferation of the endothelium is a highly coordinated process to ensure the emergence, expansion, and homeostasis of the vasculature. While Bone Morphogenetic Protein (BMP) signaling fine-tunes the behaviors of endothelium in health and disease, how BMP signaling influences the proliferation of endothelium and therefore, modulates angiogenesis remains largely unknown. Here, we evaluated the role of Activin A Type I Receptor (ACVR1/ALK2), a key BMP receptor in the endothelium, in modulating the proliferation of endothelial cells. We show that ACVR1/ALK2 is a key modulator for the proliferation of endothelium in the retinal vessels. Loss of endothelial ALK2 leads to a significant reduction in endothelial proliferation and results in fewer branches/endothelial cells in the retinal vessels. Interestingly, venous endothelium appears to be more susceptible to ALK2 deletion. Mechanistically, ACVR1/ALK2 inhibits the expression of CDKN1A/p21, a critical negative regulator of cell cycle progression, in a SMAD1/5-dependent manner, thereby enabling the venous endothelium to undergo active proliferation by suppressing CDKN1A/p21. Taken together, our findings show that BMP signaling mediated by ACVR1/ALK2 provides a critical yet previously underappreciated input to modulate the proliferation of venous endothelium, thereby fine-tuning the context of angiogenesis in health and disease.
Collapse
Affiliation(s)
- Boryeong Pak
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Minjung Kim
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Orjin Han
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Heon-Woo Lee
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Pharmacy, Chosun University, Gwangju, Korea
| | - Alexandre Dubrac
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- CHU Sainte-Justine Research Center, and Department of Pathology and Cellular Biology, Université de Montréal, Montréal, QC, Canada
| | - Woosoung Choi
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Jee Myung Yang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Kevin Boyé
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Heewon Cho
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Kathryn M Citrin
- Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Injune Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Anne Eichmann
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Victoria L Bautch
- Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Suk-Won Jin
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea.
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
27
|
Repczynska A, Ciastek B, Haus O. New Insights into the Fanconi Anemia Pathogenesis: A Crosstalk Between Inflammation and Oxidative Stress. Int J Mol Sci 2024; 25:11619. [PMID: 39519169 PMCID: PMC11547024 DOI: 10.3390/ijms252111619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Fanconi anemia (FA) represents a rare hereditary disease; it develops due to germline pathogenic variants in any of the 22 currently discovered FANC genes, which interact with the Fanconi anemia/breast cancer-associated (FANC/BRCA) pathway to maintain genome integrity. FA is characterized by a triad of clinical traits, including congenital anomalies, bone marrow failure (BMF) and multiple cancer susceptibility. Due to the complex genetic background and a broad spectrum of FA clinical symptoms, the diagnostic process is complex and requires the use of classical cytogenetic, molecular cytogenetics and strictly molecular methods. Recent findings indicate the interplay of inflammation, oxidative stress, disrupted mitochondrial metabolism, and impaired intracellular signaling in the FA pathogenesis. Additionally, a shift in the balance towards overproduction of proinflammatory cytokines and prooxidant components in FA is associated with advanced myelosuppression and ultimately BMF. Although the mechanism of BMF is very complex and needs further clarification, it appears that mutual interaction between proinflammatory cytokines and redox imbalance causes pancytopenia. In this review, we summarize the available literature regarding the clinical phenotype, genetic background, and diagnostic procedures of FA. We also highlight the current understanding of disrupted autophagy process, proinflammatory state, impaired signaling pathways and oxidative genotoxic stress in FA pathogenesis.
Collapse
Affiliation(s)
- Anna Repczynska
- Department of Clinical Genetics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Curie Sklodowskiej 9, 85-094 Bydgoszcz, Poland;
| | - Barbara Ciastek
- Institute of Health Sciences, University of Opole, Katowicka 68, 45-060 Opole, Poland
| | - Olga Haus
- Department of Clinical Genetics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Curie Sklodowskiej 9, 85-094 Bydgoszcz, Poland;
| |
Collapse
|
28
|
Janus P, Kuś P, Jaksik R, Vydra N, Toma-Jonik A, Gramatyka M, Kurpas M, Kimmel M, Widłak W. Transcriptional responses to direct and indirect TGFB1 stimulation in cancerous and noncancerous mammary epithelial cells. Cell Commun Signal 2024; 22:522. [PMID: 39468555 PMCID: PMC11514872 DOI: 10.1186/s12964-024-01821-5] [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: 03/26/2024] [Accepted: 09/07/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Transforming growth factor beta (TGFβ) is important for the morphogenesis and secretory function of the mammary gland. It is one of the main activators of the epithelial-mesenchymal transition (EMT), a process important for tissue remodeling and regeneration. It also provides cells with the plasticity to form metastases during tumor progression. Noncancerous and cancer cells respond differently to TGFβ. However, knowledge of the cellular signaling cascades triggered by TGFβ in various cell types is still limited. METHODS MCF10A (noncancerous, originating from fibrotic breast tissue) and MCF7 (cancer, estrogen receptor-positive) breast epithelial cells were treated with TGFB1 directly or through conditioned media from stimulated cells. Transcriptional changes (via RNA-seq) were assessed in untreated cells and after 1-6 days of treatment. Differentially expressed genes were detected with DESeq2 and the hallmark collection was selected for gene set enrichment analysis. RESULTS TGFB1 induces EMT in both the MCF10A and MCF7 cell lines but via slightly different mechanisms (signaling through SMAD3 is more active in MCF7 cells). Many EMT-related genes are expressed in MCF10A cells at baseline. Both cell lines respond to TGFB1 by decreasing the expression of genes involved in cell proliferation: through the repression of MYC (and the protein targets) in MCF10A cells and the activation of p63-dependent signaling in MCF7 cells (CDKN1A and CDKN2B, which are responsible for the inhibition of cyclin-dependent kinases, are upregulated). In addition, estrogen receptor signaling is inhibited and caspase-dependent cell death is induced only in MCF7 cells. Direct incubation with TGFB1 and treatment of cells with conditioned media similarly affected transcriptional profiles. However, TGFB1-induced protein secretion is more pronounced in MCF10A cells; therefore, the signaling is propagated through conditioned media (bystander effect) more effectively in MCF10A cells than in MCF7 cells. CONCLUSIONS Estrogen receptor-positive breast cancer patients may benefit from high levels of TGFB1 expression due to the repression of estrogen receptor signaling, inhibition of proliferation, and induction of apoptosis in cancer cells. However, some TGFB1-stimulated cells may undergo EMT, which increases the risk of metastasis.
Collapse
Affiliation(s)
- Patryk Janus
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland
| | - Paweł Kuś
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland
| | - Roman Jaksik
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland
| | - Natalia Vydra
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland
| | - Agnieszka Toma-Jonik
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland
| | - Michalina Gramatyka
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland
| | - Monika Kurpas
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland
| | - Marek Kimmel
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland.
- Departments of Statistics and Bioengineering, Rice University, Houston, TX, USA.
| | - Wiesława Widłak
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland.
| |
Collapse
|
29
|
Wong YS, Mançanares AC, Navarrete F, Poblete P, Mendez-Pérez L, Rodriguez-Alvarez L, Castro FO. Short preconditioning with TGFβ of equine adipose tissue-derived mesenchymal stem cells predisposes towards an anti-fibrotic secretory phenotype: A possible tool for treatment of endometrosis in mares. Theriogenology 2024; 225:119-129. [PMID: 38805994 DOI: 10.1016/j.theriogenology.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/30/2024]
Abstract
Endometrosis in mares is a disease resulting from chronic inflammation characterized by peri glandular fibrosis. There is no effective treatment so far, which opens the door for exploring the use of stem cells as a candidate. Transforming growth factor beta (TGFβ) is crucial for the establishment and progression of fibrosis in mare's endometrosis. We aimed to develop regenerative approaches to treat endometrosis by using mesenchymal stem cells (MSC), for which understanding the effect of TGFβ on exogenous MSC is crucial. We isolated and characterized equine adipose MSC from six donors. Cells were pooled and exposed to 10 ng/ml of TGFβ for 0, 4, and 24 h, after which cells were analyzed for proliferation, migration, mesodermal differentiation, expression of fibrosis-related mRNAs, and prostaglandin E2 secretion. At 24 h of exposition to TGFβ, there was a progressive increase in the contraction of the monolayer, leading to nodular structures, while cell viability did not change. Exposure to TGFβ impaired adipogenic and osteogenic differentiation after 4 h of treatment, which was more marked at 24 h, represented by a decrease in Oil red and Alizarin red staining, as well as a significant drop (p < 0.05) in the expression of key gene regulators of differentiation processes (PPARG for adipose and RUNX2 for osteogenic differentiation). TGFβ increased chondrogenic differentiation as shown by the upsurge in size of the resulting 3D cell pellet and intensity of Alcian Blue staining, as well as the significant up-regulation of SOX9 expression (p < 0.05) at 4 h, which reached a maximum peak at 24 h (p < 0.01), indicative of up-regulation of glycosaminoglycan synthesis. Preconditioning MSC with TGFβ led to a significant increase (p < 0.05) in the expression of myofibroblast gene markers aSMA, COL1A1, and TGFβ at 24 h exposition time. In contrast, the expression of COL3A1 did not change with respect to the control but registered a significant downregulation compared to 4 h (p < 0.05). TGFβ also affected the expression of genes involved in PGE2 synthesis and function; COX2, PTGES, and the PGE2 receptor EP4 were all significantly upregulated early at 4 h (p < 0.05). Cells exposed to TGFβ showed a significant upregulation of PGE2 secretion at 4 h compared to untreated cells (p < 0.05); conversely, at 24 h, the PGE2 values decreased significantly compared to control cells (p < 0.05). Preconditioning MSC for 4 h led to an anti-fibrotic secretory phenotype, while a longer period (24 h) led to a pro-fibrotic one. It is tempting to propose a 4-h preconditioning of exogenous MSC with TGFβ to drive them towards an anti-fibrotic phenotype for cellular and cell-free therapies in fibrotic diseases such as endometrosis of mares.
Collapse
Affiliation(s)
- Yat Sen Wong
- Ph.D Program in Veterinary Sciences, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| | - Ana Carolina Mançanares
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| | - Felipe Navarrete
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| | - Pamela Poblete
- Ph.D Program in Veterinary Sciences, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| | - Lidice Mendez-Pérez
- Ph.D Program in Veterinary Sciences, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| | | | - Fidel Ovidio Castro
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile.
| |
Collapse
|
30
|
Zelisko N, Lesyk R, Stoika R. Structure, unique biological properties, and mechanisms of action of transforming growth factor β. Bioorg Chem 2024; 150:107611. [PMID: 38964148 DOI: 10.1016/j.bioorg.2024.107611] [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: 01/21/2024] [Revised: 06/07/2024] [Accepted: 06/30/2024] [Indexed: 07/06/2024]
Abstract
Transforming growth factor β (TGF-β) is a ubiquitous molecule that is extremely conserved structurally and plays a systemic role in human organism. TGF-β is a homodimeric molecule consisting of two subunits joined through a disulphide bond. In mammals, three genes code for TGF-β1, TGF-β2, and TGF-β3 isoforms of this cytokine with a dominating expression of TGF-β1. Virtually, all normal cells contain TGF-β and its specific receptors. Considering the exceptional role of fine balance played by the TGF-β in anumber of physiological and pathological processes in human body, this cytokine may be proposed for use in medicine as an immunosuppressant in transplantology, wound healing and bone repair. TGFb itself is an important target in oncology. Strategies for blocking members of TGF-β signaling pathway as therapeutic targets have been considered. In this review, signalling mechanisms of TGF-β1 action are addressed, and their role in physiology and pathology with main focus on carcinogenesis are described.
Collapse
Affiliation(s)
- Nataliya Zelisko
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine.
| | - Rostyslav Stoika
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine
| |
Collapse
|
31
|
Wang X, Zhang L, Si H. Combining luteolin and curcumin synergistically suppresses triple-negative breast cancer by regulating IFN and TGF-β signaling pathways. Biomed Pharmacother 2024; 178:117221. [PMID: 39111078 DOI: 10.1016/j.biopha.2024.117221] [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: 02/22/2024] [Revised: 07/15/2024] [Accepted: 07/26/2024] [Indexed: 08/25/2024] Open
Abstract
Combining two or more chemicals in chemotherapy is rapidly increasing because of its higher efficacy, lower toxicity, lower dosages, and lower drug resistance. Here, we identified a novel combination of luteolin (LUT) and curcumin (CUR), two bioactive compounds from foods, synergistically suppressed triple-negative breast cancer (TNBC) cell proliferation (LUT 30 µM + CUR 20 µM), colony formation (LUT 1 µM + CUR 2 µM), and tumor growth in xenograft mice (LUT 10 mg/kg body weight/day + CUR 20 mg/kg body weight/day, i.p. injection every other day, 5 weeks), while the individual chemical alone did not show these inhibitory effects significantly at the selected concentrations/dosages. Our total RNA transcriptome analysis in xenograft tumors revealed that combining LUT and CUR synergistically activated type I interferon (IFN) signaling and suppressed transforming growth factor-beta (TGF-β) signaling pathways, which was further confirmed by the expression/activity of several proteins of the pathways in tumors. In addition, this combination of LUT and CUR also synergistically decreased oncoprotein levels of c-Myc and Notch1, the critical molecules required to maintain stem cell properties, tumor clonal evolution, and drug resistance. These results suggest that the combination of LUT and CUR synergistically inhibits TNBC by suppressing multiple cellular mechanisms, such as proliferation, colony formation, and transformation, as well as tumor migration, invasion, and metastasis, via regulating IFN and TGF-β signaling pathways. Therefore, combining LUT and CUR may be an effective therapeutic agent to treat highly aggressive, drug-resistant TNBC patients after clinical trials.
Collapse
Affiliation(s)
- Xiaoyong Wang
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN 37209, USA; Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lijuan Zhang
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Hongwei Si
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN 37209, USA.
| |
Collapse
|
32
|
Němec V, Remeš M, Beňovský P, Böck MC, Šranková E, Wong JF, Cros J, Williams E, Tse LH, Smil D, Ensan D, Isaac MB, Al-Awar R, Gomolková R, Ursachi VC, Fafílek B, Kahounová Z, Víchová R, Vacek O, Berger BT, Wells CI, Corona CR, Vasta JD, Robers MB, Krejci P, Souček K, Bullock AN, Knapp S, Paruch K. Discovery of Two Highly Selective Structurally Orthogonal Chemical Probes for Activin Receptor-like Kinases 1 and 2. J Med Chem 2024; 67:12632-12659. [PMID: 39023313 PMCID: PMC11320582 DOI: 10.1021/acs.jmedchem.4c00629] [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: 03/18/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024]
Abstract
Activin receptor-like kinases 1-7 (ALK1-7) regulate a complex network of SMAD-independent as well as SMAD-dependent signaling pathways. One of the widely used inhibitors for functional investigations of these processes, in particular for bone morphogenetic protein (BMP) signaling, is LDN-193189. However, LDN-193189 has insufficient kinome-wide selectivity complicating its use in cellular target validation assays. Herein, we report the identification and comprehensive characterization of two chemically distinct highly selective inhibitors of ALK1 and ALK2, M4K2234 and MU1700, along with their negative controls. We show that both MU1700 and M4K2234 efficiently block the BMP pathway via selective in cellulo inhibition of ALK1/2 kinases and exhibit favorable in vivo profiles in mice. MU1700 is highly brain penetrant and shows remarkably high accumulation in the brain. These high-quality orthogonal chemical probes offer the selectivity required to become widely used tools for in vitro and in vivo investigation of BMP signaling.
Collapse
Affiliation(s)
- Václav Němec
- Institute
for Pharmaceutical Chemistry, Structural Genomics Consortium, Johann Wolfgang Goethe-University, Max-von-Laue-Strasse 9, Frankfurt am Main, 60438, Germany
- Department
of Chemistry, Masaryk University, Brno 625 00, Czech Republic
| | - Marek Remeš
- Department
of Chemistry, Masaryk University, Brno 625 00, Czech Republic
| | - Petr Beňovský
- Department
of Chemistry, Masaryk University, Brno 625 00, Czech Republic
| | - Michael C. Böck
- Department
of Chemistry, Masaryk University, Brno 625 00, Czech Republic
| | - Eliška Šranková
- Department
of Chemistry, Masaryk University, Brno 625 00, Czech Republic
| | - Jong Fu Wong
- Centre
for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K.
| | - Julien Cros
- Centre
for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K.
| | - Eleanor Williams
- Centre
for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K.
| | - Lap Hang Tse
- Centre
for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K.
| | - David Smil
- Drug
Discovery Program, Ontario Institute for
Cancer Research, 661 University Avenue, Toronto, Ontario M5G 0A3, Canada
| | - Deeba Ensan
- Drug
Discovery Program, Ontario Institute for
Cancer Research, 661 University Avenue, Toronto, Ontario M5G 0A3, Canada
| | - Methvin B. Isaac
- Drug
Discovery Program, Ontario Institute for
Cancer Research, 661 University Avenue, Toronto, Ontario M5G 0A3, Canada
| | - Rima Al-Awar
- Drug
Discovery Program, Ontario Institute for
Cancer Research, 661 University Avenue, Toronto, Ontario M5G 0A3, Canada
- Department
of Pharmacology and Toxicology, University
of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Regina Gomolková
- Department
of Biology, Faculty of Medicine, Masaryk
University, 625 00 Brno, Czech
Republic
- Institute
of Animal Physiology and Genetics of the Czech Academy of Sciences, 602 00 Brno, Czech Republic
| | - Vlad-Constantin Ursachi
- Department
of Biology, Faculty of Medicine, Masaryk
University, 625 00 Brno, Czech
Republic
- International
Clinical Research Center, St. Anne’s
University Hospital, 602
00 Brno, Czech Republic
| | - Bohumil Fafílek
- Department
of Biology, Faculty of Medicine, Masaryk
University, 625 00 Brno, Czech
Republic
- Institute
of Animal Physiology and Genetics of the Czech Academy of Sciences, 602 00 Brno, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital, 602
00 Brno, Czech Republic
| | - Zuzana Kahounová
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 00 Brno Czech Republic
| | - Ráchel Víchová
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 00 Brno Czech Republic
| | - Ondřej Vacek
- International
Clinical Research Center, St. Anne’s
University Hospital, 602
00 Brno, Czech Republic
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 00 Brno Czech Republic
| | - Benedict-Tilman Berger
- Institute
for Pharmaceutical Chemistry, Structural Genomics Consortium, Johann Wolfgang Goethe-University, Max-von-Laue-Strasse 9, Frankfurt am Main, 60438, Germany
| | - Carrow I. Wells
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | | | - James D. Vasta
- Promega Corporation, Madison, Wisconsin 53716, United States
| | | | - Pavel Krejci
- Department
of Biology, Faculty of Medicine, Masaryk
University, 625 00 Brno, Czech
Republic
- Institute
of Animal Physiology and Genetics of the Czech Academy of Sciences, 602 00 Brno, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital, 602
00 Brno, Czech Republic
| | - Karel Souček
- International
Clinical Research Center, St. Anne’s
University Hospital, 602
00 Brno, Czech Republic
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 00 Brno Czech Republic
| | - Alex N. Bullock
- Centre
for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, U.K.
| | - Stefan Knapp
- Institute
for Pharmaceutical Chemistry, Structural Genomics Consortium, Johann Wolfgang Goethe-University, Max-von-Laue-Strasse 9, Frankfurt am Main, 60438, Germany
| | - Kamil Paruch
- Department
of Chemistry, Masaryk University, Brno 625 00, Czech Republic
- Institute
of Animal Physiology and Genetics of the Czech Academy of Sciences, 602 00 Brno, Czech Republic
| |
Collapse
|
33
|
Cheng YC, Zhang Y, Tripathi S, Harshavardhan BV, Jolly MK, Schiebinger G, Levine H, McDonald TO, Michor F. Reconstruction of single-cell lineage trajectories and identification of diversity in fates during the epithelial-to-mesenchymal transition. Proc Natl Acad Sci U S A 2024; 121:e2406842121. [PMID: 39093947 PMCID: PMC11317558 DOI: 10.1073/pnas.2406842121] [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: 04/10/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Exploring the complexity of the epithelial-to-mesenchymal transition (EMT) unveils a diversity of potential cell fates; however, the exact timing and mechanisms by which early cell states diverge into distinct EMT trajectories remain unclear. Studying these EMT trajectories through single-cell RNA sequencing is challenging due to the necessity of sacrificing cells for each measurement. In this study, we employed optimal-transport analysis to reconstruct the past trajectories of different cell fates during TGF-beta-induced EMT in the MCF10A cell line. Our analysis revealed three distinct trajectories leading to low EMT, partial EMT, and high EMT states. Cells along the partial EMT trajectory showed substantial variations in the EMT signature and exhibited pronounced stemness. Throughout this EMT trajectory, we observed a consistent downregulation of the EED and EZH2 genes. This finding was validated by recent inhibitor screens of EMT regulators and CRISPR screen studies. Moreover, we applied our analysis of early-phase differential gene expression to gene sets associated with stemness and proliferation, pinpointing ITGB4, LAMA3, and LAMB3 as genes differentially expressed in the initial stages of the partial versus high EMT trajectories. We also found that CENPF, CKS1B, and MKI67 showed significant upregulation in the high EMT trajectory. While the first group of genes aligns with findings from previous studies, our work uniquely pinpoints the precise timing of these upregulations. Finally, the identification of the latter group of genes sheds light on potential cell cycle targets for modulating EMT trajectories.
Collapse
Affiliation(s)
- Yu-Chen Cheng
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA02215
- Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
| | - Yun Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100021, China
| | - Shubham Tripathi
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale School of Medicine, New Haven, CT06510
| | - B. V. Harshavardhan
- Interdisciplinary Mathematics Initiative, Indian Institute of Science, Bangalore560012, India
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore560012, India
| | - Geoffrey Schiebinger
- Department of Mathematics, University of British Columbia, Vancouver, BCV6T 1Z2, Canada
| | - Herbert Levine
- Center for Theoretical Biological Physics, Northeastern University, Boston, MA02115
- Department of Physics, Northeastern University, Boston, MA02115
| | - Thomas O. McDonald
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA02215
- Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
| | - Franziska Michor
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA02215
- Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02138
- The Ludwig Center at Harvard, Boston, MA02115
| |
Collapse
|
34
|
Wang L, Wang Z, Ni Y, Wang X, Zhang T, Hu M, Lian C, Wang X, Zhang J. Elucidating the mechanism of action of Isobavachalcone induced autophagy and apoptosis in non-small cell lung cancer by network pharmacology and experimental validation methods. Gene 2024; 918:148474. [PMID: 38670393 DOI: 10.1016/j.gene.2024.148474] [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: 12/09/2023] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Lung cancer is the leading cause of cancer deaths, and non-small cell lung cancer (NSCLC) accounts for the majority of lung cancer-related mortality. In recent years, there have been numerous treatments for non-small cell lung cancer, but the cure and survival rates are still extremely low. Isobavachalcone (IBC) belongs to the chalcone component of the traditional Chinese medicine Psoralea corylifolia L., and is a unique Protein kinase B (AKT) pathway inhibitor with significant anticancer effects. Previous studies have shown that IBC possess a variety of biological properties, including anti-cancer, anti-inflammatory, and antioxidant properties. This study focused on the use of network pharmacology analysis, molecular docking technology and experimental validation to elucidate the potential mechanisms of IBC for the treatment of NSCLC. METHODS Screening key genes and pathways of IBC action in NSCLC using network pharmacology. The IBC target genes were from The Encyclopedia of Traditional Chinese Medicine (ETCM) and BATMAN-TCM databases, the NSCLC target genes were from GeneCards, Online Mendelian Inheritance in Man (OMIM) and The Therapeutic Target database (TTD) databases, both of which were taken as intersecting genes for protein-protein interaction network analysis and enrichment analysis, and the binding energies of the compounds to the core targets were further verified by molecular docking. Cell lines in vitro experiments were then performed to further unravel the mechanism of IBC for NSCLC. RESULTS A total of 279 potential targets were retrieved by searching the intersection of IBC and NSCLC targets. Protein-protein interaction (PPI) network analysis indicated that 6 targets, including AKT1, RXRA, NCOA1, RXRB, RARA, PPARG were hub genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis suggested that IBC treatment of NSCLC mainly involves steroid binding, transcription factor activity, Pathways in cancer, cAMP signaling pathway, Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling pathway. Among them, the AMPK signaling pathway, which contained the largest number of enriched genes, may play a greater role in the treatment of NSCLC. Then, the results of in vitro experiment indicated that IBC could inhibit proliferation of NSCLC cells and induce cell autophagy and apoptosis. The results also showed that IBC could increase the protein expression of AMPK and decrease the protein expression of AKT and mammalian target of rapamycin (mTOR), suggesting that IBC can treat NSCLC by inducing cellular autophagy and apoptosis as well as modulating AMPK and AKT signaling pathways. CONCLUSIONS In summary, this study provided a new insight into the protective mechanism of IBC against NSCLC through network pharmacology and experimental validation.
Collapse
Affiliation(s)
- Luyao Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233030, China; Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu 233030, China
| | - Ziqiang Wang
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu 233030, China
| | - Yuhan Ni
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233030, China
| | - Xue Wang
- Digestive Department, Xi'an Fifth Hospital, Xi'an 710000, China
| | - Tingting Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu 233030, China
| | - Mengling Hu
- Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu 233030, China
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu 233030, China.
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233030, China; Joint Research Center for Regional Diseases of IHM, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233030, China.
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu 233030, China.
| |
Collapse
|
35
|
Zhao H, Zhao H, Ji S. A Mesenchymal stem cell Aging Framework, from Mechanisms to Strategies. Stem Cell Rev Rep 2024; 20:1420-1440. [PMID: 38727878 DOI: 10.1007/s12015-024-10732-4] [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] [Accepted: 05/02/2024] [Indexed: 08/13/2024]
Abstract
Mesenchymal stem cells (MSCs) are extensively researched for therapeutic applications in tissue engineering and show significant potential for clinical use. Intrinsic or extrinsic factors causing senescence may lead to reduced proliferation, aberrant differentiation, weakened immunoregulation, and increased inflammation, ultimately limiting the potential of MSCs. It is crucial to comprehend the molecular pathways and internal processes responsible for the decline in MSC function due to senescence in order to devise innovative approaches for rejuvenating senescent MSCs and enhancing MSC treatment. We investigate the main molecular processes involved in senescence, aiming to provide a thorough understanding of senescence-related issues in MSCs. Additionally, we analyze the most recent advancements in cutting-edge approaches to combat MSC senescence based on current research. We are curious whether the aging process of stem cells results in a permanent "memory" and if cellular reprogramming may potentially revert the aging epigenome to a more youthful state.
Collapse
Affiliation(s)
- Hongqing Zhao
- Nanbu County People's Hospital, Nanchong City, 637300, Sichuan Province, China
- Jinzhou Medical University, No.82 Songpo Road, Guta District, Jinzhou, 121001, Liaoning Province, China
| | - Houming Zhao
- Graduate School of PLA Medical College, Chinese PLA General Hospital, Beijing, 100083, China
| | - Shuaifei Ji
- Graduate School of PLA Medical College, Chinese PLA General Hospital, Beijing, 100083, China.
| |
Collapse
|
36
|
Duan S, Yang Q, Wu F, Li Z, Hong W, Cao M, Chen X, Zhong X, Zhou Q, Zhao H. Maternal methylosome protein 50 is essential for embryonic development in medaka Oryzias latipes. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2024; 341:798-810. [PMID: 38654580 DOI: 10.1002/jez.2824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/06/2024] [Accepted: 03/20/2024] [Indexed: 04/26/2024]
Abstract
Methylosome protein 50 (Mep50) is a protein that is rich in WD40 domains, which mediate and regulate a variety of physiological processes in organisms. Previous studies indicated the necessity of Mep50 in embryogenesis in mice Mus musculus and fish. This study aimed to further understand the roles of maternal Mep50 in early embryogenesis using medaka Oryzias latipes as a model. Without maternal Mep50, medaka zygotes developed to the pre-early gastrula stage but died later. The transcriptome of the embryos at the pre-early gastrula stage was analyzed by RNA sequencing. The results indicated that 1572 genes were significantly upregulated and 741 genes were significantly downregulated in the embryos without maternal Mep50. In the differentially expressed genes (DEGs), the DNA-binding proteins, such as histones and members of the small chromosome maintenance complex, were enriched. The major interfered regulatory networks in the embryos losing maternal Mep50 included DNA replication and cell cycle regulation, AP-1 transcription factors such as Jun and Fos, the Wnt pathway, RNA processing, and the extracellular matrix. Quantitative RT-PCR verified 16 DEGs, including prmt5, H2A, cpsf, jun, mcm4, myc, p21, ccne2, cdk6, and col1, among others. It was speculated that the absence of maternal Mep50 could potentially lead to errors in DNA replication and cell cycle arrest, ultimately resulting in cell apoptosis. This eventually resulted in the failure of gastrulation and embryonic death. The results indicate the importance of maternal Mep50 in early embryonic development, particularly in medaka fish.
Collapse
Affiliation(s)
- Shi Duan
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Qing Yang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Fan Wu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Zhenyu Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Wentao Hong
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Mengxi Cao
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan, China
| | - Xinhua Chen
- Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xueping Zhong
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Qingchun Zhou
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Haobin Zhao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| |
Collapse
|
37
|
Lahane GP, Dhar A, Bhat A. Therapeutic approaches and novel antifibrotic agents in renal fibrosis: A comprehensive review. J Biochem Mol Toxicol 2024; 38:e23795. [PMID: 39132761 DOI: 10.1002/jbt.23795] [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: 01/25/2024] [Revised: 06/20/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024]
Abstract
Renal fibrosis (RF) is one of the underlying pathological conditions leading to progressive loss of renal function and end-stage renal disease (ESRD). Over the years, various therapeutic approaches have been explored to combat RF and prevent ESRD. Despite significant advances in understanding the underlying molecular mechanism(s), effective therapeutic interventions for RF are limited. Current therapeutic strategies primarily target these underlying mechanisms to halt or reverse fibrotic progression. Inhibition of transforming growth factor-β (TGF-β) signaling, a pivotal mediator of RF has emerged as a central strategy to manage RF. Small molecules, peptides, and monoclonal antibodies that target TGF-β receptors or downstream effectors have demonstrated potential in preclinical models. Modulating the renin-angiotensin system and targeting the endothelin system also provide established approaches for controlling fibrosis-related hemodynamic changes. Complementary to pharmacological strategies, lifestyle modifications, and dietary interventions contribute to holistic management. This comprehensive review aims to summarize the underlying mechanisms of RF and provide an overview of the therapeutic strategies and novel antifibrotic agents that hold promise in its treatment.
Collapse
Affiliation(s)
- Ganesh Panditrao Lahane
- Department of Pharmacy, Birla Institute of Technology and Sciences (BITS) Pilani, Hyderabad, Telangana, India
| | - Arti Dhar
- Department of Pharmacy, Birla Institute of Technology and Sciences (BITS) Pilani, Hyderabad, Telangana, India
| | - Audesh Bhat
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu and Kashmir, India
| |
Collapse
|
38
|
Szeőcs D, Vida B, Petővári G, Póliska S, Janka E, Sipos A, Uray K, Sebestyén A, Krasznai Z, Bai P. Cell-free ascites from ovarian cancer patients induces Warburg metabolism and cell proliferation through TGFβ-ERK signaling. GeroScience 2024; 46:3581-3597. [PMID: 38196068 PMCID: PMC11226691 DOI: 10.1007/s11357-023-01056-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/24/2023] [Indexed: 01/11/2024] Open
Abstract
Ascites plays a key role in supporting the metastatic potential of ovarian cancer cells. Shear stress and carry-over of cancer cells by ascites flow support carcinogenesis and metastasis formation. In addition, soluble factors may participate in the procarcinogenic effects of ascites in ovarian cancer. This study aimed to determine the biological effects of cell-free ascites on carcinogenesis in ovarian cancer cells. Cell-free ascites from ovarian cancer patients (ASC) non-selectively induced cell proliferation in multiple models of ovarian cancer and untransformed primary human dermal fibroblasts. Furthermore, ASC induced a Warburg-type rearrangement of cellular metabolism in A2780 ovarian cancer cells characterized by increases in cellular oxygen consumption and glycolytic flux; increases in glycolytic flux were dominant. ASC induced mitochondrial uncoupling and fundamentally reduced fatty acid oxidation. Ascites-elicited effects were uniform among ascites specimens. ASC-elicited transcriptomic changes in A2780 ovarian cancer cells included induction of the TGFβ-ERK/MEK pathway, which plays a key role in inducing cell proliferation and oncometabolism. ASC-induced gene expression changes, as well as the overexpression of members of the TGFβ signaling system, were associated with poor survival in ovarian cancer patients. We provided evidence that the activation of the autocrine/paracrine of TGFβ signaling system may be present in bladder urothelial carcinoma and stomach adenocarcinoma. Database analysis suggests that the TGFβ system may feed forward bladder urothelial carcinoma and stomach adenocarcinoma. Soluble components of ASC support the progression of ovarian cancer. These results suggest that reducing ascites production may play an essential role in the treatment of ovarian cancer by inhibiting the progression and reducing the severity of the disease.
Collapse
Affiliation(s)
- Dóra Szeőcs
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary, 4032
- Center of Excellence, The Hungarian Academy of Sciences, Debrecen, Hungary
| | - Beáta Vida
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary, 4032
| | - Gábor Petővári
- Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Szilárd Póliska
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary, 4032
| | - Eszter Janka
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary, 4032
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary, 4032
- Center of Excellence, The Hungarian Academy of Sciences, Debrecen, Hungary
- HUN-REN-DE Cell Biology and Signaling Research Group, Debrecen, Hungary, 4032
| | - Karen Uray
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary, 4032
- Center of Excellence, The Hungarian Academy of Sciences, Debrecen, Hungary
| | - Anna Sebestyén
- Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Zoárd Krasznai
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary, 4032
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary, 4032.
- Center of Excellence, The Hungarian Academy of Sciences, Debrecen, Hungary.
- HUN-REN-DE Cell Biology and Signaling Research Group, Debrecen, Hungary, 4032.
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary, 4032.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary, 4032.
| |
Collapse
|
39
|
Larionov A, Hammer CM, Fiedler K, Filgueira L. Dynamics of Endothelial Cell Diversity and Plasticity in Health and Disease. Cells 2024; 13:1276. [PMID: 39120307 PMCID: PMC11312403 DOI: 10.3390/cells13151276] [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: 06/27/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Endothelial cells (ECs) are vital structural units of the cardiovascular system possessing two principal distinctive properties: heterogeneity and plasticity. Endothelial heterogeneity is defined by differences in tissue-specific endothelial phenotypes and their high predisposition to modification along the length of the vascular bed. This aspect of heterogeneity is closely associated with plasticity, the ability of ECs to adapt to environmental cues through the mobilization of genetic, molecular, and structural alterations. The specific endothelial cytoarchitectonics facilitate a quick structural cell reorganization and, furthermore, easy adaptation to the extrinsic and intrinsic environmental stimuli, known as the epigenetic landscape. ECs, as universally distributed and ubiquitous cells of the human body, play a role that extends far beyond their structural function in the cardiovascular system. They play a crucial role in terms of barrier function, cell-to-cell communication, and a myriad of physiological and pathologic processes. These include development, ontogenesis, disease initiation, and progression, as well as growth, regeneration, and repair. Despite substantial progress in the understanding of endothelial cell biology, the role of ECs in healthy conditions and pathologies remains a fascinating area of exploration. This review aims to summarize knowledge and concepts in endothelial biology. It focuses on the development and functional characteristics of endothelial cells in health and pathological conditions, with a particular emphasis on endothelial phenotypic and functional heterogeneity.
Collapse
Affiliation(s)
- Alexey Larionov
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Christian Manfred Hammer
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Klaus Fiedler
- Independent Researcher, CH-1700 Fribourg, Switzerland;
| | - Luis Filgueira
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| |
Collapse
|
40
|
Thomas R, Jerome JM, Krieger KL, Ashraf N, Rowley DR. The reactive stroma response regulates the immune landscape in prostate cancer. JOURNAL OF TRANSLATIONAL GENETICS AND GENOMICS 2024; 8:249-77. [DOI: 10.20517/jtgg.2024.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Prostate cancer remains the most commonly diagnosed and the second leading cause of cancer-related deaths in men in the United States. The neoplastic transformation of prostate epithelia, concomitant with modulations in the stromal compartment, known as reactive stromal response, is critical for the growth, development, and progression of prostate cancer. Reactive stroma typifies an emergent response to disrupted tissue homeostasis commonly observed in wound repair and pathological conditions such as cancer. Despite the significance of reactive stroma in prostate cancer pathobiology, our understanding of the ontogeny, phenotypic and functional heterogeneity, and reactive stromal regulation of the immune microenvironment in prostate cancer remains limited. Traditionally characterized to have an immunologically "cold" tumor microenvironment, prostate cancer presents significant challenges for advancing immunotherapy compared to other solid tumors. This review explores the detrimental role of reactive stroma in prostate cancer, particularly its immunomodulatory function. Understanding the molecular characteristics and dynamic transcriptional program of the reactive stromal populations in tandem with tumor progression could offer insights into enhancing immunotherapy efficacy against prostate cancer.
Collapse
|
41
|
Mustafa M, Abbas K, Alam M, Habib S, Zulfareen, Hasan GM, Islam S, Shamsi A, Hassan I. Investigating underlying molecular mechanisms, signaling pathways, emerging therapeutic approaches in pancreatic cancer. Front Oncol 2024; 14:1427802. [PMID: 39087024 PMCID: PMC11288929 DOI: 10.3389/fonc.2024.1427802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Pancreatic adenocarcinoma, a clinically challenging malignancy constitutes a significant contributor to cancer-related mortality, characterized by an inherently poor prognosis. This review aims to provide a comprehensive understanding of pancreatic adenocarcinoma by examining its multifaceted etiologies, including genetic mutations and environmental factors. The review explains the complex molecular mechanisms underlying its pathogenesis and summarizes current therapeutic strategies, including surgery, chemotherapy, and emerging modalities such as immunotherapy. Critical molecular pathways driving pancreatic cancer development, including KRAS, Notch, and Hedgehog, are discussed. Current therapeutic strategies, including surgery, chemotherapy, and radiation, are discussed, with an emphasis on their limitations, particularly in terms of postoperative relapse. Promising research areas, including liquid biopsies, personalized medicine, and gene editing, are explored, demonstrating the significant potential for enhancing diagnosis and treatment. While immunotherapy presents promising prospects, it faces challenges related to immune evasion mechanisms. Emerging research directions, encompassing liquid biopsies, personalized medicine, CRISPR/Cas9 genome editing, and computational intelligence applications, hold promise for refining diagnostic approaches and therapeutic interventions. By integrating insights from genetic, molecular, and clinical research, innovative strategies that improve patient outcomes can be developed. Ongoing research in these emerging fields holds significant promise for advancing the diagnosis and treatment of this formidable malignancy.
Collapse
Affiliation(s)
- Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Zulfareen
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sidra Islam
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates
| | - Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
42
|
Li H, Wang G, Zhao G, Liu H, Liu L, Cao Y, Li C. TGF-β1 maintains the developmental potential of embryonic submandibular gland epithelia separated with mesenchyme. Heliyon 2024; 10:e33506. [PMID: 39040362 PMCID: PMC11261778 DOI: 10.1016/j.heliyon.2024.e33506] [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: 03/06/2023] [Revised: 06/11/2024] [Accepted: 06/21/2024] [Indexed: 07/24/2024] Open
Abstract
Objective The objective of this study was to investigate the impact of transforming growth factor β1 (TGF-β1) on epithelial development using an ex vivo model of submandibular gland (SMG) epithelial-mesenchymal separation. Materials and methods The ex vivo model was established by separating E13 mouse SMG epithelia and mesenchyme, culturing them independently for 24 h, recombining them, and observing branching morphogenesis. Microarray analysis was performed to evaluate the transcriptome of epithelia treated with and without 1 ng/ml TGF-β1. Differential gene expression, pathway enrichment, and protein-protein interaction networks were analyzed. Quantitative real-time polymerase chain reaction, Western blot, and immunofluorescence were employed to validate the mRNA and protein levels. Results Recombined SMGs using separated epithelia and mesenchyme that were cultured for 24 h showed a significant inhibition of epithelial development compared to SMGs recombined immediately after separation. The level of TGF-β1 decreased in the SMG epithelia after epithelia-mesenchyme separation. Epithelia that were separated from mesenchyme for 24 h and pretreated with 1 ng/ml TGF-β1 continued to develop after recombination with mesenchyme, while epithelia without 1 ng/ml TGF-β1 treatment did not. Microarray analysis suggested pathway enrichment related to epithelial development and an upregulation of Sox2 in the 1 ng/ml TGF-β1-treated epithelia. Further experiments validated the phosphorylation of SMAD2 and SMAD3, upregulation of SOX2 and genes associated with epithelial development, including Prol1, Dcpp1, Bhlha15, Smgc, and Bpifa2. Additionally, 1 ng/ml TGF-β1 inhibited epithelial apoptosis by improving the BCL2/BAX ratio and reducing cleaved caspase 3. Conclusions The addition of 1 ng/ml TGF-β1 maintained the developmental potential of embryonic SMG epithelia separated from mesenchyme for 24 h. This suggests that 1 ng/ml TGF-β1 may partially compensate for the role of mesenchyme during the separation phase, although its compensation is limited in extent.
Collapse
Affiliation(s)
- Honglin Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Guanru Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Guile Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Huabing Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Liu Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yubin Cao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chunjie Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
43
|
Gu M, Wang Y, Yu Y. Ovarian fibrosis: molecular mechanisms and potential therapeutic targets. J Ovarian Res 2024; 17:139. [PMID: 38970048 PMCID: PMC11225137 DOI: 10.1186/s13048-024-01448-7] [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: 05/11/2024] [Accepted: 06/03/2024] [Indexed: 07/07/2024] Open
Abstract
Ovarian fibrosis, characterized by the excessive proliferation of ovarian fibroblasts and the accumulation of extracellular matrix (ECM), serves as one of the primary causes of ovarian dysfunction. Despite the critical role of ovarian fibrosis in maintaining the normal physiological function of the mammalian ovaries, research on this condition has been greatly underestimated, which leads to a lack of clinical treatment options for ovarian dysfunction caused by fibrosis. This review synthesizes recent research on the molecular mechanisms of ovarian fibrosis, encompassing TGF-β, extracellular matrix, inflammation, and other profibrotic factors contributing to abnormal ovarian fibrosis. Additionally, we summarize current treatment approaches for ovarian dysfunction targeting ovarian fibrosis, including antifibrotic drugs, stem cell transplantation, and exosomal therapies. The purpose of this review is to summarize the research progress on ovarian fibrosis and to propose potential therapeutic strategies targeting ovarian fibrosis for the treatment of ovarian dysfunction.
Collapse
Affiliation(s)
- Mengqing Gu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Ministry of Education, Beijing, 100191, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Yibo Wang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction (Peking University), Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Ministry of Education, Beijing, 100191, China.
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China.
- Institute of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Yang Yu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction (Peking University), Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Ministry of Education, Beijing, 100191, China.
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| |
Collapse
|
44
|
Moskova-Doumanova V, Vaseva A, Veleva R, Mladenova K, Melniska D, Doumanov J, Videv P, Topouzova-Hristova T, Dobreva L, Atanasova N, Danova S. In Vitro Effects of Postmetabolites from Limosilactobacillus fermentum 53 on the Survival and Proliferation of HT-29 Cells. Microorganisms 2024; 12:1365. [PMID: 39065133 PMCID: PMC11279320 DOI: 10.3390/microorganisms12071365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/13/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Naturally fermented dairy products are an important component of the human diet. They are a valuable source of nutrients as well as vitamins and minerals. Their importance as a source of probiotic bacterial strains should not be overlooked. A number of studies highlight the positive effects of species of the probiotic lactic acid bacteria on the intestinal microbiome and the overall homeostasis of the body, as well as a complementary treatment for some diseases. However, data on the effects on the intestinal epithelial cells of postmetabolites released by probiotic bacteria are incomplete. This is likely due to the fact that these effects are species- and strain-specific. In the present study, we investigated the effects of postmetabolites produced by a pre-selected candidate probiotic strain Limosilactobacillus fermentum on HT-29 intestinal epithelial cells. Our data showed a pronounced proliferative effect, evaluated by flow cytometry, quantification of the cell population and determination of the mitotic index. This was accompanied by the stabilization of the cell monolayer, measured by an increase in TEER (transepithelial electric resistance) and the reorganization of actin filaments. The data obtained are a clear indication of the positive effects that the products secreted by L. fermentum strain 53 have on intestinal epithelial cells.
Collapse
Affiliation(s)
- Veselina Moskova-Doumanova
- Faculty of Biology, Department of Cell and Developmental Biology, Sofia University St. Kliment Ohridski, 8 Dragan Tsankov Blvd., 1164 Sofia, Bulgaria; (V.M.-D.); (T.T.-H.)
| | - Anita Vaseva
- Faculty of Biology, Department of Cell and Developmental Biology, Sofia University St. Kliment Ohridski, 8 Dragan Tsankov Blvd., 1164 Sofia, Bulgaria; (V.M.-D.); (T.T.-H.)
| | - Ralitsa Veleva
- Faculty of Biology, Department of Cell and Developmental Biology, Sofia University St. Kliment Ohridski, 8 Dragan Tsankov Blvd., 1164 Sofia, Bulgaria; (V.M.-D.); (T.T.-H.)
| | - Kirilka Mladenova
- Faculty of Biology, Department of Biochemistry, Sofia University St. Kliment Ohridski, 8 Dragan Tsankov Blvd., 1164 Sofia, Bulgaria; (K.M.); (J.D.)
| | - Denitsa Melniska
- Faculty of Biology, Department of Cell and Developmental Biology, Sofia University St. Kliment Ohridski, 8 Dragan Tsankov Blvd., 1164 Sofia, Bulgaria; (V.M.-D.); (T.T.-H.)
| | - Jordan Doumanov
- Faculty of Biology, Department of Biochemistry, Sofia University St. Kliment Ohridski, 8 Dragan Tsankov Blvd., 1164 Sofia, Bulgaria; (K.M.); (J.D.)
| | - Pavel Videv
- Faculty of Biology, Department of Biochemistry, Sofia University St. Kliment Ohridski, 8 Dragan Tsankov Blvd., 1164 Sofia, Bulgaria; (K.M.); (J.D.)
| | - Tanya Topouzova-Hristova
- Faculty of Biology, Department of Cell and Developmental Biology, Sofia University St. Kliment Ohridski, 8 Dragan Tsankov Blvd., 1164 Sofia, Bulgaria; (V.M.-D.); (T.T.-H.)
| | - Lili Dobreva
- Bulgarian Academy of Sciences, Stefan Angeloff Institute of Microbiology, 26, Acad. Georgi Bonchev Street, 1113 Sofia, Bulgaria; (L.D.); (N.A.)
| | - Nikoleta Atanasova
- Bulgarian Academy of Sciences, Stefan Angeloff Institute of Microbiology, 26, Acad. Georgi Bonchev Street, 1113 Sofia, Bulgaria; (L.D.); (N.A.)
| | - Svetla Danova
- Bulgarian Academy of Sciences, Stefan Angeloff Institute of Microbiology, 26, Acad. Georgi Bonchev Street, 1113 Sofia, Bulgaria; (L.D.); (N.A.)
| |
Collapse
|
45
|
Li D, Liu C, Wang H, Li Y, Wang Y, An S, Sun S. The Role of Neuromodulation and Potential Mechanism in Regulating Heterotopic Ossification. Neurochem Res 2024; 49:1628-1642. [PMID: 38416374 DOI: 10.1007/s11064-024-04118-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/17/2024] [Accepted: 01/28/2024] [Indexed: 02/29/2024]
Abstract
Heterotopic ossification (HO) is a pathological process characterized by the aberrant formation of bone in muscles and soft tissues. It is commonly triggered by traumatic brain injury, spinal cord injury, and burns. Despite a wide range of evidence underscoring the significance of neurogenic signals in proper bone remodeling, a clear understanding of HO induced by nerve injury remains rudimentary. Recent studies suggest that injury to the nervous system can activate various signaling pathways, such as TGF-β, leading to neurogenic HO through the release of neurotrophins. These pathophysiological changes lay a robust groundwork for the prevention and treatment of HO. In this review, we collected evidence to elucidate the mechanisms underlying the pathogenesis of HO related to nerve injury, aiming to enhance our understanding of how neurological repair processes can culminate in HO.
Collapse
Affiliation(s)
- Dengju Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong First Medical University, Jinan, Shandong, China
| | - Changxing Liu
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Haojue Wang
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yunfeng Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yaqi Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Senbo An
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong First Medical University, Jinan, Shandong, China.
| | - Shui Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
46
|
Yoshimoto S, Yada N, Ishikawa A, Kawano K, Matsuo K, Hiraki A, Okamura K. TGF-β3 from fibroblasts promotes necrotising sialometaplasia by suppressing salivary gland cell proliferation and inducing squamous metaplasia. J Pathol 2024; 263:338-346. [PMID: 38594209 DOI: 10.1002/path.6287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/25/2024] [Accepted: 03/20/2024] [Indexed: 04/11/2024]
Abstract
Necrotising sialometaplasia (NSM) is a non-neoplastic lesion mainly arising in the minor salivary glands of the oral cavity. In the clinical features, NSM shows swelling with or without ulceration, and can mimic a malignant disease such as squamous cell carcinoma. Histopathologically, NSM usually shows the lobular architecture that is observed in the salivary glands. Additionally, acinar infarction and squamous metaplasia of salivary ducts and acini are observable. The aetiology of this lesion remains unknown, although it has a characteristic feature that sometimes requires clinical and histopathological differentiation from malignancy. In this study, we investigated upregulated genes in NSM compared with normal salivary glands, and focused on the TGF-β3 (TGFB3) gene. The results of the histopathological studies clarified that fibroblasts surrounding the lesion express TGF-β3. Moreover, in vitro studies using mouse salivary gland organoids revealed that TGF-β3 suppressed salivary gland cell proliferation and induced squamous metaplasia. We demonstrated a possible aetiology of NSM by concluding that increased TGF-β3 expression during wound healing or tissue regeneration played a critical role in cell proliferation and metaplasia. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Shohei Yoshimoto
- Section of Pathology, Department of Morphological Biology, Division of Biomedical Sciences, Fukuoka Dental College, Fukuoka, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Naomi Yada
- Department of Health Promotion, Division of Oral Pathology, Kyushu Dental University, Kitakyushu, Japan
| | | | - Kenji Kawano
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Kou Matsuo
- Department of Health Promotion, Division of Oral Pathology, Kyushu Dental University, Kitakyushu, Japan
| | - Akimitsu Hiraki
- Section of Oral Oncology, Department of Oral and Maxillofacial Surgery, Division of Oral and Medical Management, Fukuoka Dental College, Fukuoka, Japan
| | - Kazuhiko Okamura
- Section of Pathology, Department of Morphological Biology, Division of Biomedical Sciences, Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
47
|
Hou Y, Wang H, Wu J, Guo H, Chen X. Dissecting the pleiotropic roles of reactive oxygen species (ROS) in lung cancer: From carcinogenesis toward therapy. Med Res Rev 2024; 44:1566-1595. [PMID: 38284170 DOI: 10.1002/med.22018] [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: 08/23/2022] [Revised: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 01/30/2024]
Abstract
Lung cancer is a major cause of morbidity and mortality. The specific pulmonary structure to directly connect with ambient air makes it more susceptible to damage from airborne toxins. External oxidative stimuli and endogenous reactive oxygen species (ROS) play a crucial role in promoting lung carcinogenesis and development. The biological properties of higher ROS levels in tumor cells than in normal cells make them more sensitive and vulnerable to ROS injury. Therefore, the strategy of targeting ROS has been proposed for cancer therapy for decades. However, it is embarrassing that countless attempts at ROS-based therapies have had very limited success, and no FDA approval in the anticancer list was mechanistically based on ROS manipulation. Even compared with the untargetable proteins, such as transcription factors, ROS are more difficult to be targeted due to their chemical properties. Thus, the pleiotropic roles of ROS provide therapeutic potential for anticancer drug discovery, while a better dissection of the mechanistic action and signaling pathways is a prerequisite for future breakthroughs. This review discusses the critical roles of ROS in cancer carcinogenesis, ROS-inspired signaling pathways, and ROS-based treatment, exemplified by lung cancer. In particular, an eight considerations rule is proposed for ROS-targeting strategies and drug design and development.
Collapse
Affiliation(s)
- Ying Hou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Heng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Hongwei Guo
- Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
- Department of Pharmaceutical Sciences, University of Macau, Taipa, Macao, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao, China
| |
Collapse
|
48
|
Lan XQ, Deng CJ, Wang QQ, Zhao LM, Jiao BW, Xiang Y. The role of TGF-β signaling in muscle atrophy, sarcopenia and cancer cachexia. Gen Comp Endocrinol 2024; 353:114513. [PMID: 38604437 DOI: 10.1016/j.ygcen.2024.114513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/24/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Skeletal muscle, comprising a significant proportion (40 to 50 percent) of total body weight in humans, plays a critical role in maintaining normal physiological conditions. Muscle atrophy occurs when the rate of protein degradation exceeds protein synthesis. Sarcopenia refers to age-related muscle atrophy, while cachexia represents a more complex form of muscle wasting associated with various diseases such as cancer, heart failure, and AIDS. Recent research has highlighted the involvement of signaling pathways, including IGF1-Akt-mTOR, MuRF1-MAFbx, and FOXO, in regulating the delicate balance between muscle protein synthesis and breakdown. Myostatin, a member of the TGF-β superfamily, negatively regulates muscle growth and promotes muscle atrophy by activating Smad2 and Smad3. It also interacts with other signaling pathways in cachexia and sarcopenia. Inhibition of myostatin has emerged as a promising therapeutic approach for sarcopenia and cachexia. Additionally, other TGF-β family members, such as TGF-β1, activin A, and GDF11, have been implicated in the regulation of skeletal muscle mass. Furthermore, myostatin cooperates with these family members to impair muscle differentiation and contribute to muscle loss. This review provides an overview of the significance of myostatin and other TGF-β signaling pathway members in muscular dystrophy, sarcopenia, and cachexia. It also discusses potential novel therapeutic strategies targeting myostatin and TGF-β signaling for the treatment of muscle atrophy.
Collapse
Affiliation(s)
- Xin-Qiang Lan
- Metabolic Control and Aging Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Cheng-Jie Deng
- Department of Biochemistry and Molecular Biology, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Qi-Quan Wang
- Metabolic Control and Aging Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Li-Min Zhao
- Senescence and Cancer Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Bao-Wei Jiao
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Yang Xiang
- Metabolic Control and Aging Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China.
| |
Collapse
|
49
|
Song Y, Gu D, Gao N, Sa H, Wang R, Fang L, Yuan Z. Smad4 deficiency inhibits lung metastases through enhancing phagocytosis of lung interstitial macrophages. Biochem Biophys Res Commun 2024; 715:150007. [PMID: 38678783 DOI: 10.1016/j.bbrc.2024.150007] [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: 04/09/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
Smad4, a critical mediator of TGF-β signaling, plays a pivotal role in regulating various cellular functions, including immune responses. In this study, we investigated the impact of Smad4 knockout specifically in macrophages on anti-tumor immunity, focusing on lung metastasis of B16 melanoma cells. Using a mouse model with Smad4 knockout in macrophages established via Lyz2-cre mice and Smad4 flox/flox mice, we demonstrated a significant inhibition of B16 metastasis in the lungs. Interestingly, the inhibition of tumor growth was found to be independent of adaptive immunity, as no significant changes were observed in the numbers or activities of T cells, B cells, or NK cells. Instead, Smad4 knockout led to the emergence of an MCHIIlow CD206high subset of lung interstitial macrophages, characterized by enhanced phagocytosis function. Our findings highlight the crucial role of Smad4 in modulating the innate immune response against tumors and provide insights into potential therapeutic strategies targeting lung interstitial macrophages to enhance anti-tumor immunity.
Collapse
Affiliation(s)
- Yu Song
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China.
| | - Dongxu Gu
- Department of Transfusion Medicine, The Third Bethune Hospital of Jilin University, 2, Xiantai Street, Changchun, 130012, China.
| | - Nan Gao
- Laboratory Department, The Third Affiliated Hospital of CCUCM, 1643, Jingyue Street, Changchun, 130021, China.
| | - Huanlan Sa
- Cancer Center, The First Hospital of Jilin University, 1, Xinmin Street, Changchun, 130021, China.
| | - Ruonan Wang
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China.
| | - Lin Fang
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China.
| | - Zhaoxin Yuan
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China.
| |
Collapse
|
50
|
Gois LL, Ribeiro-Soares B, Regis-Silva CG, Zanette DL, Lisboa R, Nascimento RS, Coutinho Junior R, Galvão-Castro B, Grassi MFR. Imbalanced IL10/TGF-β production by regulatory T-lymphocytes in patients with HTLV-1-associated myelopathy/ tropical spastic paraparesis. BMC Infect Dis 2024; 24:652. [PMID: 38943078 PMCID: PMC11214226 DOI: 10.1186/s12879-024-09494-8] [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: 12/21/2023] [Accepted: 06/11/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND Human T-cell lymphotropic virus type 1 (HTLV-1), also denominated Human T-cell leukemia virus-1, induces immune activation and secretion of proinflammatory cytokines, especially in individuals with HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Regulatory T lymphocytes (Tregs) may control of inflammation through the production of regulatory cytokines, including IL10 and TGF-β. In this study we determined the frequencies of CD4 + and CD8 + Tregs in a HAM/TSP population, compared to asymptomatic carriers and uninfected individuals, as well as investigated the profiles of regulatory and inflammatory cytokines. METHODS Asymptomatic HTLV-1 carriers and HAM/TSP patients were matched by sex and age. The frequencies of IL10- and/or TGF-β-producing Tregs were quantified by flow cytometry. Real-time reverse transcription polymerase chain reaction (RT-PCR) was used to quantify HTLV-1 proviral load and the mRNA expression of cytokines and cellular receptors in peripheral blood mononuclear cells. RESULTS Total frequencies of CD4 + Tregs, as well as the IL10-producing CD4 + and CD8 + Treg subsets, were statistically higher in patients with HAM/TSP compared to asymptomatic HTLV-1-infected individuals. In addition, a positive correlation was found between the frequency of CD4 + IL10 + Tregs and proviral load in the HAM/TSP patients evaluated. A positive correlation was also observed between gene expression of proinflammatory versus regulatory cytokines only in HAM / TSP group. CONCLUSIONS A higher frequencies of IL10-producing Tregs were identified in patients with HAM/TSP. Imbalanced production of IL10 in relation to TGF-β may contribute to the increased inflammatory response characteristically seen in HAM/TSP patients.
Collapse
Affiliation(s)
- Luana Leandro Gois
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador-Bahia, Brazil
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (LASP, IGM, FIOCRUZ), Salvador-Bahia, Brazil
- Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia (ICS/UFBA), Salvador-Bahia, Brazil
| | - Bárbara Ribeiro-Soares
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador-Bahia, Brazil
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (LASP, IGM, FIOCRUZ), Salvador-Bahia, Brazil
| | - Carlos Gustavo Regis-Silva
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (LASP, IGM, FIOCRUZ), Salvador-Bahia, Brazil
| | - Dalila L Zanette
- Laboratório de Ciências e Tecnologias Aplicadas a Saúde, Instituto Carlos Chagas, Fundação Oswaldo Cruz (ICC/FIOCRUZ-PR), Curitiba-Paraná, Brazil
| | - Raphaella Lisboa
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador-Bahia, Brazil
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (LASP, IGM, FIOCRUZ), Salvador-Bahia, Brazil
| | - Regina Santos Nascimento
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (LASP, IGM, FIOCRUZ), Salvador-Bahia, Brazil
| | - Raimundo Coutinho Junior
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador-Bahia, Brazil
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (LASP, IGM, FIOCRUZ), Salvador-Bahia, Brazil
| | - Bernardo Galvão-Castro
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador-Bahia, Brazil
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (LASP, IGM, FIOCRUZ), Salvador-Bahia, Brazil
| | - Maria Fernanda Rios Grassi
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador-Bahia, Brazil.
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (LASP, IGM, FIOCRUZ), Salvador-Bahia, Brazil.
| |
Collapse
|