1
|
Yuasa-Kawada J, Kinoshita-Kawada M, Hiramoto M, Yamagishi S, Mishima T, Yasunaga S, Tsuboi Y, Hattori N, Wu JY. Neuronal guidance signaling in neurodegenerative diseases: Key regulators that function at neuron-glia and neuroimmune interfaces. Neural Regen Res 2026; 21:612-635. [PMID: 39995079 DOI: 10.4103/nrr.nrr-d-24-01330] [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: 10/31/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
The nervous system processes a vast amount of information, performing computations that underlie perception, cognition, and behavior. During development, neuronal guidance genes, which encode extracellular cues, their receptors, and downstream signal transducers, organize neural wiring to generate the complex architecture of the nervous system. It is now evident that many of these neuroguidance cues and their receptors are active during development and are also expressed in the adult nervous system. This suggests that neuronal guidance pathways are critical not only for neural wiring but also for ongoing function and maintenance of the mature nervous system. Supporting this view, these pathways continue to regulate synaptic connectivity, plasticity, and remodeling, and overall brain homeostasis throughout adulthood. Genetic and transcriptomic analyses have further revealed many neuronal guidance genes to be associated with a wide range of neurodegenerative and neuropsychiatric disorders. Although the precise mechanisms by which aberrant neuronal guidance signaling drives the pathogenesis of these diseases remain to be clarified, emerging evidence points to several common themes, including dysfunction in neurons, microglia, astrocytes, and endothelial cells, along with dysregulation of neuron-microglia-astrocyte, neuroimmune, and neurovascular interactions. In this review, we explore recent advances in understanding the molecular and cellular mechanisms by which aberrant neuronal guidance signaling contributes to disease pathogenesis through altered cell-cell interactions. For instance, recent studies have unveiled two distinct semaphorin-plexin signaling pathways that affect microglial activation and neuroinflammation. We discuss the challenges ahead, along with the therapeutic potentials of targeting neuronal guidance pathways for treating neurodegenerative diseases. Particular focus is placed on how neuronal guidance mechanisms control neuron-glia and neuroimmune interactions and modulate microglial function under physiological and pathological conditions. Specifically, we examine the crosstalk between neuronal guidance signaling and TREM2, a master regulator of microglial function, in the context of pathogenic protein aggregates. It is well-established that age is a major risk factor for neurodegeneration. Future research should address how aging and neuronal guidance signaling interact to influence an individual's susceptibility to various late-onset neurological diseases and how the progression of these diseases could be therapeutically blocked by targeting neuronal guidance pathways.
Collapse
Affiliation(s)
| | | | | | - Satoru Yamagishi
- Department of Optical Neuroanatomy, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takayasu Mishima
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Japan
| | - Shin'ichiro Yasunaga
- Department of Biochemistry, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jane Y Wu
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Duan Y, Yang F, Zhang Y, Zhang M, Shi Y, Lang Y, Sun H, Wang X, Jin H, Kang X. Role of mitophagy in spinal cord ischemia-reperfusion injury. Neural Regen Res 2026; 21:598-611. [PMID: 39665804 DOI: 10.4103/nrr.nrr-d-24-00668] [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: 06/18/2024] [Accepted: 10/29/2024] [Indexed: 12/13/2024] Open
Abstract
Spinal cord ischemia-reperfusion injury, a severe form of spinal cord damage, can lead to sensory and motor dysfunction. This injury often occurs after traumatic events, spinal cord surgeries, or thoracoabdominal aortic surgeries. The unpredictable nature of this condition, combined with limited treatment options, poses a significant burden on patients, their families, and society. Spinal cord ischemia-reperfusion injury leads to reduced neuronal regenerative capacity and complex pathological processes. In contrast, mitophagy is crucial for degrading damaged mitochondria, thereby supporting neuronal metabolism and energy supply. However, while moderate mitophagy can be beneficial in the context of spinal cord ischemia-reperfusion injury, excessive mitophagy may be detrimental. Therefore, this review aims to investigate the potential mechanisms and regulators of mitophagy involved in the pathological processes of spinal cord ischemia-reperfusion injury. The goal is to provide a comprehensive understanding of recent advancements in mitophagy related to spinal cord ischemia-reperfusion injury and clarify its potential clinical applications.
Collapse
Affiliation(s)
- Yanni Duan
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Fengguang Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yibao Zhang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Mingtao Zhang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yujun Shi
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yun Lang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Hongli Sun
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Xin Wang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Hongyun Jin
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Xuewen Kang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| |
Collapse
|
3
|
Wen J, Tang H, Tian M, Wang L, Yang Q, Zhao Y, Li X, Ren Y, Wang J, Zhou L, Tan Y, Wu H, Cai X, Wang Y, Cao H, Xu J, Yang Q. Fibrotic scar formation after cerebral ischemic stroke: Targeting the Sonic hedgehog signaling pathway for scar reduction. Neural Regen Res 2026; 21:756-768. [PMID: 40183351 DOI: 10.4103/nrr.nrr-d-24-00999] [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: 08/28/2024] [Accepted: 12/30/2024] [Indexed: 04/05/2025] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00044/figure1/v/2025-05-05T160104Z/r/image-tiff Recent studies have shown that fibrotic scar formation following cerebral ischemic injury has varying effects depending on the microenvironment. However, little is known about how fibrosis is induced and regulated after cerebral ischemic injury. Sonic hedgehog signaling participates in fibrosis in the heart, liver, lung, and kidney. Whether Shh signaling modulates fibrotic scar formation after cerebral ischemic stroke and the underlying mechanisms are unclear. In this study, we found that Sonic Hedgehog expression was upregulated in patients with acute ischemic stroke and in a middle cerebral artery occlusion/reperfusion injury rat model. Both Sonic hedgehog and Mitofusin 2 showed increased expression in the middle cerebral artery occlusion rat model and in vitro fibrosis cell model induced by transforming growth factor-beta 1. Activation of the Sonic hedgehog signaling pathway enhanced the expression of phosphorylated Smad 3 and Mitofusin 2 proteins, promoted the formation of fibrotic scars, protected synapses or promoted synaptogenesis, alleviated neurological deficits following middle cerebral artery occlusion/reperfusion injury, reduced cell apoptosis, facilitated the transformation of meninges fibroblasts into myofibroblasts, and enhanced the proliferation and migration of meninges fibroblasts. The Smad3 phosphorylation inhibitor SIS3 reversed the effects induced by Sonic hedgehog signaling pathway activation. Bioinformatics analysis revealed significant correlations between Sonic hedgehog and Smad3, between Sonic hedgehog and Mitofusin 2, and between Smad3 and Mitofusin 2. These findings suggest that Sonic hedgehog signaling may influence Mitofusin 2 expression by regulating Smad3 phosphorylation, thereby modulating the formation of early fibrotic scars following cerebral ischemic stroke and affecting prognosis. The Sonic Hedgehog signaling pathway may serve as a new therapeutic target for stroke treatment.
Collapse
Affiliation(s)
- Jun Wen
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Tang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingfen Tian
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Wang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinghuan Yang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Zhao
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuemei Li
- Department of Neurology, Second People's Hospital of Chongqing Banan District, Chongqing, China
| | - Yu Ren
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiani Wang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zhou
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yongjun Tan
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haiyun Wu
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinrui Cai
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yilin Wang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Cao
- Department of Neurosurgery, Third Hospital of Mianyang, Mianyang, Sichuan Province, China
| | - Jianfeng Xu
- Department of Neurosurgery, Third Hospital of Mianyang, Mianyang, Sichuan Province, China
| | - Qin Yang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Liu Z, Guo Y, Zhang Y, Gao Y, Ning B. Metabolic reprogramming of astrocytes: Emerging roles of lactate. Neural Regen Res 2026; 21:421-432. [PMID: 39688570 DOI: 10.4103/nrr.nrr-d-24-00776] [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: 07/16/2024] [Accepted: 10/25/2024] [Indexed: 12/18/2024] Open
Abstract
Lactate serves as a key energy metabolite in the central nervous system, facilitating essential brain functions, including energy supply, signaling, and epigenetic modulation. Moreover, it links epigenetic modifications with metabolic reprogramming. Nonetheless, the specific mechanisms and roles of this connection in astrocytes remain unclear. Therefore, this review aims to explore the role and specific mechanisms of lactate in the metabolic reprogramming of astrocytes in the central nervous system. The close relationship between epigenetic modifications and metabolic reprogramming was discussed. Therapeutic strategies for targeting metabolic reprogramming in astrocytes in the central nervous system were also outlined to guide future research in central nervous system diseases. In the nervous system, lactate plays an essential role. However, its mechanism of action as a bridge between metabolic reprogramming and epigenetic modifications in the nervous system requires future investigation. The involvement of lactate in epigenetic modifications is currently a hot research topic, especially in lactylation modification, a key determinant in this process. Lactate also indirectly regulates various epigenetic modifications, such as N6-methyladenosine, acetylation, ubiquitination, and phosphorylation modifications, which are closely linked to several neurological disorders. In addition, exploring the clinical applications and potential therapeutic strategies of lactic acid provides new insights for future neurological disease treatments.
Collapse
Affiliation(s)
- Zeyu Liu
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Yijian Guo
- Department of Spinal Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Ying Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Yulei Gao
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Bin Ning
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
- Department of Spinal Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
5
|
Wu S, Chen J. Is age-related myelinodegenerative change an initial risk factor of neurodegenerative diseases? Neural Regen Res 2026; 21:648-658. [PMID: 40326982 DOI: 10.4103/nrr.nrr-d-24-00848] [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: 07/29/2024] [Accepted: 11/25/2024] [Indexed: 05/07/2025] Open
Abstract
Myelination, the continuous ensheathment of neuronal axons, is a lifelong process in the nervous system that is essential for the precise, temporospatial conduction of action potentials between neurons. Myelin also provides intercellular metabolic support to axons. Even minor disruptions in the integrity of myelin can impair neural performance and increase susceptibility to neurological diseases. In fact, myelin degeneration is a well-known neuropathological condition that is associated with normal aging and several neurodegenerative diseases, including multiple sclerosis and Alzheimer's disease. In the central nervous system, compact myelin sheaths are formed by fully mature oligodendrocytes. However, the entire oligodendrocyte lineage is susceptible to changes in the biological microenvironment and other risk factors that arise as the brain ages. In addition to their well-known role in action potential propagation, oligodendrocytes also provide intercellular metabolic support to axons by transferring energy metabolites and delivering exosomes. Therefore, myelin degeneration in the aging central nervous system is a significant contributor to the development of neurodegenerative diseases. Interventions that mitigate age-related myelin degeneration can improve neurological function in aging individuals. In this review, we investigate the changes in myelin that are associated with aging and their underlying mechanisms. We also discuss recent advances in understanding how myelin degeneration in the aging brain contributes to neurodegenerative diseases and explore the factors that can prevent, slow down, or even reverse age-related myelin degeneration. Future research will enhance our understanding of how reducing age-related myelin degeneration can be used as a therapeutic target for delaying or preventing neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuangchan Wu
- Sanhang Institute for Brain Science and Technology (SiBST), School of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi Province, China
- Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, Guangdong Province, China
| | - Jun Chen
- Sanhang Institute for Brain Science and Technology (SiBST), School of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi Province, China
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
6
|
Yao L, Cai X, Yang S, Song Y, Xing L, Li G, Cui Z, Chen J. A single-cell landscape of the regenerating spinal cord of zebrafish. Neural Regen Res 2026; 21:780-789. [PMID: 40326988 DOI: 10.4103/nrr.nrr-d-24-01163] [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: 09/28/2024] [Accepted: 03/03/2025] [Indexed: 05/07/2025] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00046/figure1/v/2025-05-05T160104Z/r/image-tiff Unlike mammals, zebrafish possess a remarkable ability to regenerate their spinal cord after injury, making them an ideal vertebrate model for studying regeneration. While previous research has identified key cell types involved in this process, the underlying molecular and cellular mechanisms remain largely unexplored. In this study, we used single-cell RNA sequencing to profile distinct cell populations at different stages of spinal cord injury in zebrafish. Our analysis revealed that multiple subpopulations of neurons showed persistent activation of genes associated with axonal regeneration post injury, while molecular signals promoting growth cone collapse were inhibited. Radial glial cells exhibited significant proliferation and differentiation potential post injury, indicating their intrinsic roles in promoting neurogenesis and axonal regeneration, respectively. Additionally, we found that inflammatory factors rapidly decreased in the early stages following spinal cord injury, creating a microenvironment permissive for tissue repair and regeneration. Furthermore, oligodendrocytes lost maturity markers while exhibiting increased proliferation following injury. These findings demonstrated that the rapid and orderly regulation of inflammation, as well as the efficient proliferation and redifferentiation of new neurons and glial cells, enabled zebrafish to reconstruct the spinal cord. This research provides new insights into the cellular transitions and molecular programs that drive spinal cord regeneration, offering promising avenues for future research and therapeutic strategies.
Collapse
Affiliation(s)
- Lei Yao
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu Province, China
| | - Xinyi Cai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province, China
| | - Saishuai Yang
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu Province, China
| | - Yixing Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province, China
| | - Guicai Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province, China
| | - Zhiming Cui
- Department of Spine Surgery, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu Province, China
- Research Institute for Spine and Spinal Cord Disease of Nantong University, Nantong, Jiangsu Province, China
| | - Jiajia Chen
- Department of Spine Surgery, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu Province, China
- Research Institute for Spine and Spinal Cord Disease of Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
7
|
Negro S, Montecucco C, Rigoni M. Extra-pineal melatonin in perisynaptic Schwann cell-muscle fiber cross talk at the regenerating neuromuscular junction. Neural Regen Res 2026; 21:300-301. [PMID: 40489345 DOI: 10.4103/nrr.nrr-d-24-01136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/07/2024] [Indexed: 06/11/2025] Open
Affiliation(s)
- Samuele Negro
- Department of Biomedical Sciences, University of Padua, Padua, Italy (Negro S, Montecucco C, Rigoni M)
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padua, Padua, Italy (Negro S, Montecucco C, Rigoni M)
- CNR Institute of Neuroscience, Padua, Italy (Montecucco C)
| | - Michela Rigoni
- Department of Biomedical Sciences, University of Padua, Padua, Italy (Negro S, Montecucco C, Rigoni M)
| |
Collapse
|
8
|
Martinez-Salas E, Francisco-Velilla R. GEMIN5 and neurodevelopmental diseases: From functional insights to disease perception. Neural Regen Res 2026; 21:187-194. [PMID: 39819844 PMCID: PMC12094563 DOI: 10.4103/nrr.nrr-d-24-01010] [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: 08/30/2024] [Revised: 10/17/2024] [Accepted: 11/27/2024] [Indexed: 01/19/2025] Open
Abstract
GEMIN5 is a predominantly cytoplasmic multifunctional protein, known to be involved in recognizing snRNAs through its WD40 repeats domain placed at the N-terminus. A dimerization domain in the middle region acts as a hub for protein-protein interaction, while a non-canonical RNA-binding site is placed towards the C-terminus. The singular organization of structural domains present in GEMIN5 enables this protein to perform multiple functions through its ability to interact with distinct partners, both RNAs and proteins. This protein exerts a different role in translation regulation depending on its physiological state, such that while GEMIN5 down-regulates global RNA translation, the C-terminal half of the protein promotes translation of its mRNA. Additionally, GEMIN5 is responsible for the preferential partitioning of mRNAs into polysomes. Besides selective translation, GEMIN5 forms part of distinct ribonucleoprotein complexes, reflecting the dynamic organization of macromolecular complexes in response to internal and external signals. In accordance with its contribution to fundamental cellular processes, recent reports described clinical loss of function mutants suggesting that GEMIN5 deficiency is detrimental to cell growth and survival. Remarkably, patients carrying GEMIN5 biallelic variants suffer from neurodevelopmental delay, hypotonia, and cerebellar ataxia. Molecular analyses of individual variants, which are defective in protein dimerization, display decreased levels of ribosome association, reinforcing the involvement of the protein in translation regulation. Importantly, the number of clinical variants and the phenotypic spectrum associated with GEMIN5 disorders is increasing as the knowledge of the protein functions and the pathways linked to its activity augments. Here we discuss relevant advances concerning the functional and structural features of GEMIN5 and its separate domains in RNA-binding, protein interactome, and translation regulation, and how these data can help to understand the involvement of protein malfunction in clinical variants found in patients developing neurodevelopmental disorders.
Collapse
|
9
|
Wang B, Chen P, Li W, Chen Z. Exosomes in stroke management: A promising paradigm shift in stroke therapy. Neural Regen Res 2026; 21:6-22. [PMID: 39665811 PMCID: PMC12094539 DOI: 10.4103/nrr.nrr-d-24-00665] [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] [Revised: 07/27/2024] [Accepted: 10/31/2024] [Indexed: 12/13/2024] Open
Abstract
Effective treatment methods for stroke, a common cerebrovascular disease with a high mortality rate, are still being sought. Exosome therapy, a form of acellular therapy, has demonstrated promising efficacy in various diseases in animal models; however, there is currently insufficient evidence to guide the clinical application of exosome in patients with stroke. This article reviews the progress of exosome applications in stroke treatment. It aims to elucidate the significant potential value of exosomes in stroke therapy and provide a reference for their clinical translation. At present, many studies on exosome-based therapies for stroke are actively underway. Regarding preclinical research, exosomes, as bioactive substances with diverse sources, currently favor stem cells as their origin. Due to their high plasticity, exosomes can be effectively modified through various physical, chemical, and genetic engineering methods to enhance their efficacy. In animal models of stroke, exosome therapy can reduce neuroinflammatory responses, alleviate oxidative stress damage, and inhibit programmed cell death. Additionally, exosomes can promote angiogenesis, repair and regenerate damaged white matter fiber bundles, and facilitate the migration and differentiation of neural stem cells, aiding the repair process. We also summarize new directions for the application of exosomes, specifically the exosome intervention through the ventricular-meningeal lymphatic system. The review findings suggest that the treatment paradigm for stroke is poised for transformation.
Collapse
Affiliation(s)
- Bo Wang
- Department of Neurosurgery, The First Hospital Affiliated to Army Medical University, Chongqing, China
| | - Pinzhen Chen
- Department of Radiology, The First Hospital Affiliated to Army Medical University, Chongqing, China
| | - Wenyan Li
- Department of Neurosurgery, The First Hospital Affiliated to Army Medical University, Chongqing, China
| | - Zhi Chen
- Department of Neurosurgery, The First Hospital Affiliated to Army Medical University, Chongqing, China
| |
Collapse
|
10
|
Geng R, Wang Y, Wang R, Wu J, Bao X. Enhanced neurogenesis after ischemic stroke: The interplay between endogenous and exogenous stem cells. Neural Regen Res 2026; 21:212-223. [PMID: 39820432 PMCID: PMC12094570 DOI: 10.4103/nrr.nrr-d-24-00879] [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: 08/28/2024] [Revised: 09/02/2024] [Accepted: 11/26/2024] [Indexed: 01/19/2025] Open
Abstract
Ischemic stroke is a significant global health crisis, frequently resulting in disability or death, with limited therapeutic interventions available. Although various intrinsic reparative processes are initiated within the ischemic brain, these mechanisms are often insufficient to restore neuronal functionality. This has led to intensive investigation into the use of exogenous stem cells as a potential therapeutic option. This comprehensive review outlines the ontogeny and mechanisms of activation of endogenous neural stem cells within the adult brain following ischemic events, with focus on the impact of stem cell-based therapies on neural stem cells. Exogenous stem cells have been shown to enhance the proliferation of endogenous neural stem cells via direct cell-to-cell contact and through the secretion of growth factors and exosomes. Additionally, implanted stem cells may recruit host stem cells from their niches to the infarct area by establishing so-called "biobridges." Furthermore, xenogeneic and allogeneic stem cells can modify the microenvironment of the infarcted brain tissue through immunomodulatory and angiogenic effects, thereby supporting endogenous neuroregeneration. Given the convergence of regulatory pathways between exogenous and endogenous stem cells and the necessity for a supportive microenvironment, we discuss three strategies to simultaneously enhance the therapeutic efficacy of both cell types. These approaches include: (1) co-administration of various growth factors and pharmacological agents alongside stem cell transplantation to reduce stem cell apoptosis; (2) synergistic administration of stem cells and their exosomes to amplify paracrine effects; and (3) integration of stem cells within hydrogels, which provide a protective scaffold for the implanted cells while facilitating the regeneration of neural tissue and the reconstitution of neural circuits. This comprehensive review highlights the interactions and shared regulatory mechanisms between endogenous neural stem cells and exogenously implanted stem cells and may offer new insights for improving the efficacy of stem cell-based therapies in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ruxu Geng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuhe Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jun Wu
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| |
Collapse
|
11
|
Münz C, Campbell GR, Esclatine A, Faure M, Labonte P, Lussignol M, Orvedahl A, Altan-Bonnet N, Bartenschlager R, Beale R, Cirone M, Espert L, Jung J, Leib D, Reggiori F, Sanyal S, Spector SA, Thiel V, Viret C, Wei Y, Wileman T, Wodrich H. Autophagy machinery as exploited by viruses. AUTOPHAGY REPORTS 2025; 4:27694127.2025.2464986. [PMID: 40201908 PMCID: PMC11921968 DOI: 10.1080/27694127.2025.2464986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 04/10/2025]
Abstract
Viruses adapt and modulate cellular pathways to allow their replication in host cells. The catabolic pathway of macroautophagy, for simplicity referred to as autophagy, is no exception. In this review, we discuss anti-viral functions of both autophagy and select components of the autophagy machinery, and how viruses have evaded them. Some viruses use the membrane remodeling ability of the autophagy machinery to build their replication compartments in the cytosol or efficiently egress from cells in a non-lytic fashion. Some of the autophagy machinery components and their remodeled membranes can even be found in viral particles as envelopes or single membranes around virus packages that protect them during spreading and transmission. Therefore, studies on autophagy regulation by viral infections can reveal functions of the autophagy machinery beyond lysosomal degradation of cytosolic constituents. Furthermore, they can also pinpoint molecular interactions with which the autophagy machinery can most efficiently be manipulated, and this may be relevant to develop effective disease treatments based on autophagy modulation.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich Switzerland
| | - Grant R Campbell
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of SD, Vermillion, SD, USA
| | - Audrey Esclatine
- Université Paris-Saclay, CEA, CNRS, 10 Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Mathias Faure
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Universite Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France
| | - Patrick Labonte
- eINRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Canada
| | - Marion Lussignol
- Université Paris-Saclay, CEA, CNRS, 10 Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Anthony Orvedahl
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Nihal Altan-Bonnet
- Laboratory of Host-Pathogen Dynamics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ralf Bartenschlager
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division Virus-Associated Carcinogenesis, Heidelberg, Germany
- German Centre for Infection Research, Heidelberg partner site, Heidelberg, Germany
| | - Rupert Beale
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London, UK
- Division of Medicine, University College London, London, UK
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucile Espert
- University of Montpellier, Montpellier, France
- CNRS, Institut de Recherche enInfectiologie deMontpellier (IRIM), Montpellier, France
| | - Jae Jung
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - David Leib
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH, USA
| | - Fulvio Reggiori
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, Aarhus C, Denmark
| | - Sumana Sanyal
- Sir William Dunn School of Pathology, South Parks Road, University of Oxford, Oxford, UK
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Stephen A. Spector
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Rady Children’s Hospital, San Diego, CA, USA
| | - Volker Thiel
- Institute of Virology and Immunology, Bern and Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland, and Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Christophe Viret
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Universite Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France
| | - Yu Wei
- Institut Pasteur-Theravectys Joint Laboratory, Department of Virology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Thomas Wileman
- Norwich Medical School, University of East Anglia
- Quadram Institute Bioscience, Norwich Research Park, Norfolk, UK
| | - Harald Wodrich
- sLaboratoire de Microbiologie Fondamentale et Pathogénicité, MFP CNRS UMR, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
12
|
Ren X, Qu Y, Shari A, Li G. Transcriptome-wide study of mRNAs modified by m 6A RNA methylation in the testis development of dairy goats. Anim Biotechnol 2025; 36:2496641. [PMID: 40306318 DOI: 10.1080/10495398.2025.2496641] [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: 10/14/2024] [Accepted: 04/14/2025] [Indexed: 05/02/2025]
Abstract
N6-methyladenosine (m6A) is an important epigenetic modification in RNA, playing a crucial role in regulating the production and aging of animal testicular sperm. This study extracted mRNA from the testicular tissue of male goats before and after sexual maturity, generating a methylation map through preliminary experiments and methylation immunoprecipitation sequencing. The results showed that during the development of dairy goats, the expression levels of marker genes related to testicular development and methylation-related enzymes changed significantly. A total of 36,602 peaks and 11,223 genes were identified in the two groups, including 2989 differential peaks (427 upregulated and 2562 downregulated) and 1457 differentially expressed genes (833 upregulated and 624 downregulated). The abundance of m6A was positively correlated with gene expression levels. This study reports for the first time the mRNA profiles of m6A modifications across the entire transcriptome during testicular development in Guanzhong dairy goats, providing a new perspective for genetic improvement in goats.
Collapse
Affiliation(s)
- Xinyang Ren
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China
| | - Yingxin Qu
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China
| | - Akang Shari
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China
| | - Guang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China
| |
Collapse
|
13
|
Shan Y, Hu H, Yang A, Zhao W, Chu Y. An integrative approach to identifying NPC1 as a susceptibility gene for gestational diabetes mellitus. J Matern Fetal Neonatal Med 2025; 38:2445665. [PMID: 39746811 DOI: 10.1080/14767058.2024.2445665] [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: 04/24/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVE The objective of this study was to identify a novel gene and its potential mechanisms associated with susceptibility to gestational diabetes mellitus (GDM) through an integrative approach. METHODS We analyzed data from genome-wide association studies (GWAS) of GDM in the FinnGen R11 dataset (16,802 GDM cases and 237,816 controls) and Genotype Tissue Expression v8 expression quantitative trait locus data. We used summary-data-based Mendelian randomization to determine associations between transcript levels and phenotypes, transcriptome-wide association studies to provide insights into gene-trait associations, multi-marker analysis of genomic annotation to perform gene-based analysis, genome-wide complex trait analysis-multivariate set-based association test-combo to determine gene prioritization, and polygenic priority scores to prioritize the causal genes to screen candidate genes. Subsequent Mendelian randomization analysis was performed to infer causality between the candidate genes and GDM and phenome-wide association study (PheWAS) analysis was used to explore the associations between selected genes and other characteristics. Furthermore, to gain a deeper understanding of the functional implications of these susceptibility genes, GeneMANIA analysis was used to determine the fundamental biological functions of the therapeutic targets and protein-protein interaction network analysis was performed to identify intracellular protein interactions. RESULTS We identified two novel susceptibility genes associated with GDM: NPC1 and KIAA1191. Magnetic resonance imaging revealed a strong correlation between NPC1 expression levels and a lower incidence of GDM (odds ratio: 0.922, 95% confidence interval: 0.866-0.981, p = 0.011). PheWAS at the gene level indicated that NPC1 was not associated with any other trait. The biological significance of this gene was evidenced by its strong association with sterol metabolism. CONCLUSION Our study identified NPC1 as a novel gene whose predicted expression level is linked to a reduced risk of GDM, providing new insights into the genetic framework of this disease.
Collapse
Affiliation(s)
- Yuping Shan
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Hu
- Clinical medicine, Nantong University, Nantong, China
| | - Anning Yang
- Department of Obstetrics and Gynecology, Qingdao Eighth People's Hospital, Qingdao, China
| | - Wendi Zhao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yijing Chu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
14
|
Swain J, Askenasy I, Rudland Nazeer R, Ho PM, Labrini E, Mancini L, Xu Q, Hollendung F, Sheldon I, Dickson C, Welch A, Agbamu A, Godlee C, Welch M. Pathogenicity and virulence of Pseudomonas aeruginosa: Recent advances and under-investigated topics. Virulence 2025; 16:2503430. [PMID: 40353451 PMCID: PMC12087490 DOI: 10.1080/21505594.2025.2503430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/23/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025] Open
Abstract
Pseudomonas aeruginosa is a model for the study of quorum sensing, protein secretion, and biofilm formation. Consequently, it has become one of the most intensely reviewed pathogens, with many excellent articles in the current literature focusing on these aspects of the organism's biology. Here, though, we aim to take a slightly different approach and consider some less well appreciated (but nonetheless important) factors that affect P. aeruginosa virulence. We start by reminding the reader of the global importance of P. aeruginosa infection and that the "virulome" is very niche-specific. Overlooked but obvious questions such as "what prevents secreted protein products from being digested by co-secreted proteases?" are discussed, and we suggest how the nutritional preference(s) of the organism might dictate its environmental reservoirs. Recent studies identifying host genes associated with genetic predisposition towards P. aeruginosa infection (and even infection by specific P. aeruginosa strains) and the role(s) of intracellular P. aeruginosa are introduced. We also discuss the fact that virulence is a high-risk strategy and touch on how expression of the two main classes of virulence factors is regulated. A particular focus is on recent findings highlighting how nutritional status and metabolism are as important as quorum sensing in terms of their impact on virulence, and how co-habiting microbial species at the infection site impact on P. aeruginosa virulence (and vice versa). It is our view that investigation of these issues is likely to dominate many aspects of research into this WHO-designated priority pathogen over the next decade.
Collapse
Affiliation(s)
- Jemima Swain
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Isabel Askenasy
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | - Pok-Man Ho
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Edoardo Labrini
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | - Qingqing Xu
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | | | - Camilla Dickson
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Amelie Welch
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Adam Agbamu
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Camilla Godlee
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Martin Welch
- Department of Biochemistry, Cambridge University, Cambridge, UK
| |
Collapse
|
15
|
Choi JC. Perinuclear organelle trauma at the nexus of cardiomyopathy pathogenesis arising from loss of function LMNA mutation. Nucleus 2025; 16:2449500. [PMID: 39789731 PMCID: PMC11730615 DOI: 10.1080/19491034.2024.2449500] [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: 10/31/2024] [Revised: 12/22/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Over the past 25 years, nuclear envelope (NE) perturbations have been reported in various experimental models with mutations in the LMNA gene. Although the hypothesis that NE perturbations from LMNA mutations are a fundamental feature of striated muscle damage has garnered wide acceptance, the molecular sequalae provoked by the NE damage and how they underlie disease pathogenesis such as cardiomyopathy (LMNA cardiomyopathy) remain poorly understood. We recently shed light on one such consequence, by employing a cardiomyocyte-specific Lmna deletion in vivo in the adult heart. We observed extensive NE perturbations prior to cardiac function deterioration with collateral damage in the perinuclear space. The Golgi is particularly affected, leading to cytoprotective stress responses that are likely disrupted by the progressive deterioration of the Golgi itself. In this review, we discuss the etiology of LMNA cardiomyopathy with perinuclear 'organelle trauma' as the nexus between NE damage and disease pathogenesis.
Collapse
Affiliation(s)
- Jason C. Choi
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
16
|
Xu H, Cao L, Chen Y, Zhou C, Xu J, Zhang Z, Li X, Liu L, Lu J. Single-cell RNA sequencing reveals the heterogeneity and interactions of immune cells and Müller glia during zebrafish retina regeneration. Neural Regen Res 2025; 20:3635-3648. [PMID: 38934409 PMCID: PMC11974639 DOI: 10.4103/nrr.nrr-d-23-02083] [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: 12/26/2023] [Revised: 04/17/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202512000-00031/figure1/v/2025-01-31T122243Z/r/image-tiff Inflammation plays a crucial role in the regeneration of fish and avian retinas. However, how inflammation regulates Müller glia (MG) reprogramming remains unclear. Here, we used single-cell RNA sequencing to investigate the cell heterogeneity and interactions of MG and immune cells in the regenerating zebrafish retina. We first showed that two types of quiescent MG (resting MG1 and MG2) reside in the uninjured retina. Following retinal injury, resting MG1 transitioned into an activated state expressing known reprogramming genes, while resting MG2 gave rise to rod progenitors. We further showed that retinal microglia can be categorized into three subtypes (microglia-1, microglia-2, and proliferative) and pseudotime analysis demonstrated dynamic changes in microglial status following retinal injury. Analysis of cell-cell interactions indicated extensive crosstalk between immune cells and MG, with many interactions shared among different immune cell types. Finally, we showed that inflammation activated Jak1-Stat3 signaling in MG, promoting their transition from a resting to an activated state. Our study reveals the cell heterogeneity and crosstalk of immune cells and MG in zebrafish retinal repair, and may provide valuable insights into future mammalian retina regeneration.
Collapse
Affiliation(s)
- Hui Xu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Key Lab of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Lining Cao
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yuxi Chen
- Key Lab of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Cuiping Zhou
- Key Lab of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Jie Xu
- Key Lab of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Zhuolin Zhang
- Key Lab of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Xiangyu Li
- Key Lab of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Lihua Liu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jianfeng Lu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
17
|
Zheng Z, Lin X, Zhao Z, Lin Q, Liu J, Chen M, Wu W, Wu Z, Liu N, Chen H. A vascular endothelial growth factor-loaded chitosan-hyaluronic acid hydrogel scaffold enhances the therapeutic effect of adipose-derived stem cells in the context of stroke. Neural Regen Res 2025; 20:3591-3605. [PMID: 39248177 PMCID: PMC11974663 DOI: 10.4103/nrr.nrr-d-24-00129] [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: 01/31/2024] [Revised: 05/13/2024] [Accepted: 07/05/2024] [Indexed: 09/10/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202512000-00028/figure1/v/2025-01-31T122243Z/r/image-tiff Adipose-derived stem cell, one type of mesenchymal stem cells, is a promising approach in treating ischemia-reperfusion injury caused by occlusion of the middle cerebral artery. However, its application has been limited by the complexities of the ischemic microenvironment. Hydrogel scaffolds, which are composed of hyaluronic acid and chitosan, exhibit excellent biocompatibility and biodegradability, making them promising candidates as cell carriers. Vascular endothelial growth factor is a crucial regulatory factor for stem cells. Both hyaluronic acid and chitosan have the potential to make the microenvironment more hospitable to transplanted stem cells, thereby enhancing the therapeutic effect of mesenchymal stem cell transplantation in the context of stroke. Here, we found that vascular endothelial growth factor significantly improved the activity and paracrine function of adipose-derived stem cells. Subsequently, we developed a chitosan-hyaluronic acid hydrogel scaffold that incorporated vascular endothelial growth factor and first injected the scaffold into an animal model of cerebral ischemia-reperfusion injury. When loaded with adipose-derived stem cells, this vascular endothelial growth factor-loaded scaffold markedly reduced neuronal apoptosis caused by oxygen-glucose deprivation/reoxygenation and substantially restored mitochondrial membrane potential and axon morphology. Further in vivo experiments revealed that this vascular endothelial growth factor-loaded hydrogel scaffold facilitated the transplantation of adipose-derived stem cells, leading to a reduction in infarct volume and neuronal apoptosis in a rat model of stroke induced by transient middle cerebral artery occlusion. It also helped maintain mitochondrial integrity and axonal morphology, greatly improving rat motor function and angiogenesis. Therefore, utilizing a hydrogel scaffold loaded with vascular endothelial growth factor as a stem cell delivery system can mitigate the adverse effects of ischemic microenvironment on transplanted stem cells and enhance the therapeutic effect of stem cells in the context of stroke.
Collapse
Affiliation(s)
- Zhijian Zheng
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Xiaohui Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Zijun Zhao
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Qiang Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ji Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Manli Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Wenwen Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Zhiyun Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Nan Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Hongbin Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
18
|
Lewis EM, Becker O, Symons AN, LaCoss C, Baclig AJ, Guzman A, Sanders C, Gonzalez L, Warner LR, Lewis KA. The LARP6 La module from Tetrabaena socialis reveals structural and functional differences from plant and animal LARP6 homologues. RNA Biol 2025; 22:1-9. [PMID: 40181506 PMCID: PMC11988235 DOI: 10.1080/15476286.2025.2489303] [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: 01/24/2024] [Revised: 02/06/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025] Open
Abstract
This study identified the LARP6 La Module from Tetrabaena socialis (T. socialis), a four-celled green algae, in an effort to better understand the evolution of LARP6 structure and RNA-binding activity in multicellular eukaryotes. Using a combination of sequence alignments, domain boundary screens, and structural modelling, we recombinantly expressed and isolated the TsLARP6 La Module to > 98% purity for in vitro biochemical characterization. The La Module is stably folded and exerts minimal RNA binding activity against single-stranded homopolymeric RNAs. Surprisingly, it exhibits low micromolar binding affinity for the vertebrate LARP6 cognate ligand, a bulged-stem loop found in the 5'UTR of collagen type I mRNA, but does not bind double-stranded RNAs of similar size. These result suggests that the TsLARP6 La Module may prefer structured RNA ligands. In contrast, however, the TsLARP6 La Module does not exhibit the RNA chaperone activity that is observed in vertebrate homologs. Therefore, we conclude that protist LARP6 may have both distinct RNA ligands and binding mechanisms from the previously characterized LARP6 proteins of animals and vascular plants, thus establishing a distinct third class of the LARP6 protein family.
Collapse
Affiliation(s)
- Emily M. Lewis
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA
| | - Olga Becker
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA
| | - Alexis N. Symons
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA
| | - Cora LaCoss
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA
- San Marcos High School, San Marcos, TX, USA
| | - A. Jasmine Baclig
- Department of Chemistry and Biochemistry, Boise State University, Boise, ID, USA
| | - Avery Guzman
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA
| | - Charles Sanders
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA
| | - Leticia Gonzalez
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA
| | - Lisa R. Warner
- Department of Chemistry and Biochemistry, Boise State University, Boise, ID, USA
| | - Karen A. Lewis
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA
| |
Collapse
|
19
|
Choi H, Kwak MJ, Choi Y, Kang AN, Mun D, Eor JY, Park MR, Oh S, Kim Y. Extracellular vesicles of Limosilactobacillus fermentum SLAM216 ameliorate skin symptoms of atopic dermatitis by regulating gut microbiome on serotonin metabolism. Gut Microbes 2025; 17:2474256. [PMID: 40028723 PMCID: PMC11881872 DOI: 10.1080/19490976.2025.2474256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 12/31/2024] [Accepted: 02/25/2025] [Indexed: 03/05/2025] Open
Abstract
Atopic dermatitis (AD) is a globally prevalent chronic inflammatory skin disorder. Its pathogenesis remains incompletely understood, resulting in considerable therapeutic challenges. Recent studies have highlighted the significance of the interaction between AD and gut microbiome. In this study, we investigated the effects of probiotic-derived extracellular vesicles on AD. Initially, we isolated and characterized extracellular vesicles from Limosilactobacillus fermentum SLAM 216 (LF216EV) and characterized their composition through multi-omics analysis. Gene ontology (GO) and pathway analysis classified LF216EV proteins into biological processes, molecular functions, and cellular components. Importantly, specific abundance in linoleic, oleic, palmitic, sebacic, and stearic acids indicating upregulated fatty acid metabolism were observed by metabolomic analysis. Furthermore, featured lipid profiling including AcylGlcADG and ceramide were observed in LF216EV. Importantly, in an atopic dermatitis-like cell model induced by TNFα/IFNγ, LF216EV significantly modulated the expression of immune regulatory genes (TSLP, TNFα, IL-6, IL-1β, and MDC), indicating its potential functionality in atopic dermatitis. LF216EV alleviated AD-like phenotypes, such as redness, scaling/dryness, and excoriation, induced by DNCB. Histopathological analysis revealed that LF216EV decreased epidermal thickness and mast cell infiltration in the dermis. Furthermore, LF216EV administration reduced mouse scratching and depression-related behaviors, with a faster onset than the classical treatment with dexamethasone. In the quantitative real-time polymerase chain reaction (qRT-PCR) analysis, we observed a significant increase in the expression levels of htrb2c, sert, and tph-1, genes associated with serotonin, in the skin and gut of the LF216EV-treated group, along with a significant increase in the total serum serotonin levels. Gut microbiome analysis of the LF216EV-treated group revealed an altered gut microbiota profile. Correlation analysis revealed that the genera Limosilactobacillus and Desulfovibrio were associated with differences in the intestinal metabolites, including serotonin. Our findings demonstrate that LF216EV mitigates AD-like symptoms by promoting serotonin synthesis through the modulation of gut microbiota and metabolome composition.
Collapse
Affiliation(s)
- Hyejin Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| | - Min-Jin Kwak
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| | - Youbin Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| | - An Na Kang
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| | - Daye Mun
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| | - Ju Young Eor
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| | - Mi Ri Park
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Korea
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| |
Collapse
|
20
|
Hiruthyaswamy SP, Bose A, Upadhyay A, Raha T, Bhattacharjee S, Singha I, Ray S, Nicky Macarius NM, Viswanathan P, Deepankumar K. Molecular signaling pathways in osteoarthritis and biomaterials for cartilage regeneration: a review. Bioengineered 2025; 16:2501880. [PMID: 40336219 PMCID: PMC12064066 DOI: 10.1080/21655979.2025.2501880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/07/2025] [Accepted: 04/04/2025] [Indexed: 05/09/2025] Open
Abstract
Osteoarthritis is a prevalent degenerative joint disease characterized by cartilage degradation, synovial inflammation, and subchondral bone alterations, leading to chronic pain and joint dysfunction. Conventional treatments provide symptomatic relief but fail to halt disease progression. Recent advancements in biomaterials, molecular signaling modulation, and gene-editing technologies offer promising therapeutic strategies. This review explores key molecular pathways implicated in osteoarthritis, including fibroblast growth factor, phosphoinositide 3-kinase/Akt, and bone morphogenetic protein signaling, highlighting their roles in chondrocyte survival, extracellular matrix remodeling, and inflammation. Biomaterial-based interventions such as hydrogels, nanoparticles, and chitosan-based scaffolds have demonstrated potential in enhancing cartilage regeneration and targeted drug delivery. Furthermore, CRISPR/Cas9 gene editing holds promise in modifying osteoarthritis-related genes to restore cartilage integrity. The integration of regenerative biomaterials with precision medicine and molecular therapies represents a novel approach for mitigating osteoarthritis progression. Future research should focus on optimizing biomaterial properties, refining gene-editing efficiency, and developing personalized therapeutic strategies. The convergence of bioengineering and molecular science offers new hope for improving joint function and patient quality of life in osteoarthritis management.
Collapse
Affiliation(s)
- Samson Prince Hiruthyaswamy
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Arohi Bose
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Ayushi Upadhyay
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Tiasa Raha
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Shangomitra Bhattacharjee
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Isheeta Singha
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Swati Ray
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | | | - Pragasam Viswanathan
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Kanagavel Deepankumar
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
21
|
Nair A, Khanna J, Kler J, Ragesh R, Sengupta K. Nuclear envelope and chromatin choreography direct cellular differentiation. Nucleus 2025; 16:2449520. [PMID: 39943681 PMCID: PMC11834525 DOI: 10.1080/19491034.2024.2449520] [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: 10/01/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 02/20/2025] Open
Abstract
The nuclear envelope plays an indispensable role in the spatiotemporal organization of chromatin and transcriptional regulation during the intricate process of cell differentiation. This review outlines the distinct regulatory networks between nuclear envelope proteins, transcription factors and epigenetic modifications in controlling the expression of cell lineage-specific genes during differentiation. Nuclear lamina with its associated nuclear envelope proteins organize heterochromatin via Lamina-Associated Domains (LADs), proximal to the nuclear periphery. Since nuclear lamina is mechanosensitive, we critically examine the impact of extracellular forces on differentiation outcomes. The nuclear envelope is spanned by nuclear pore complexes which, in addition to their central role in transport, are associated with chromatin organization. Furthermore, mutations in the nuclear envelope proteins disrupt differentiation, resulting in developmental disorders. Investigating the underlying nuclear envelope controlled regulatory mechanisms of chromatin remodelling during lineage commitment will accelerate our fundamental understanding of developmental biology and regenerative medicine.
Collapse
Affiliation(s)
- Anjitha Nair
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Jayati Khanna
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Jashan Kler
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Rohith Ragesh
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Kundan Sengupta
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| |
Collapse
|
22
|
Liang H, Zhou B, Li P, Zhang X, Zhang S, Zhang Y, Yao S, Qu S, Chen J. Stemness regulation in prostate cancer: prostate cancer stem cells and targeted therapy. Ann Med 2025; 57:2442067. [PMID: 39711287 DOI: 10.1080/07853890.2024.2442067] [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: 11/27/2023] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Increasing evidence indicates that cancer stem cells (CSCs) and cancer stem-like cells form a special subpopulation of cells that are ubiquitous in tumors. These cells exhibit similar characteristics to those of normal stem cells in tissues; moreover, they are capable of self-renewal and differentiation, as well as high tumorigenicity and drug resistance. In prostate cancer (PCa), it is difficult to kill these cells using androgen signaling inhibitors and chemotherapy drugs. Consequently, the residual prostate cancer stem cells (PCSCs) mediate tumor recurrence and progression. OBJECTIVE This review aims to provide a comprehensive and up-to-date overview of PCSCs, with a particular emphasis on potential therapeutic strategies targeting these cells. METHODS After searching in PubMed and Embase databases using 'prostate cancer' and 'cancer stem cells' as keywords, studies related were compiled and examined. RESULTS In this review, we detail the origin and characteristics of PCSCs, introduce the regulatory pathways closely related to CSC survival and stemness maintenance, and discuss the link between epithelial-mesenchymal transition, tumor microenvironment and tumor stemness. Furthermore, we introduce the currently available therapeutic strategies targeting CSCs, including signaling pathway inhibitors, anti-apoptotic protein inhibitors, microRNAs, nanomedicine, and immunotherapy. Lastly, we summarize the limitations of current CSC research and mention future research directions. CONCLUSION A deeper understanding of the regulatory network and molecular markers of PCSCs could facilitate the development of novel therapeutic strategies targeting these cells. Previous preclinical studies have demonstrated the potential of this treatment approach. In the future, this may offer alternative treatment options for PCa patients.
Collapse
Affiliation(s)
- Hao Liang
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Bin Zhou
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Peixin Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyi Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shijie Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Yaozhong Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shengwen Yao
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Sifeng Qu
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Jun Chen
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| |
Collapse
|
23
|
Cong R, Lu C, Li X, Xu Z, Wang Y, Sun S. Tumor organoids in cancer medicine: from model systems to natural compound screening. PHARMACEUTICAL BIOLOGY 2025; 63:89-109. [PMID: 39893515 PMCID: PMC11789228 DOI: 10.1080/13880209.2025.2458149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/04/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025]
Abstract
CONTEXT The advent of tissue engineering and biomedical techniques has significantly advanced the development of three-dimensional (3D) cell culture systems, particularly tumor organoids. These self-assembled 3D cell clusters closely replicate the histopathological, genetic, and phenotypic characteristics of primary tissues, making them invaluable tools in cancer research and drug screening. OBJECTIVE This review addresses the challenges in developing in vitro models that accurately reflect tumor heterogeneity and explores the application of tumor organoids in cancer research, with a specific focus on the screening of natural products for antitumor therapies. METHODS This review synthesizes information from major databases, including Chemical Abstracts, Medicinal and Aromatic Plants Abstracts, ScienceDirect, Google Scholar, Scopus, PubMed and Springer Link. Publications were selected without date restrictions, using terms such as 'organoid', 'natural product', 'pharmacological', 'extract', 'nanomaterial' and 'traditional uses'. Articles related to agriculture, ecology, synthetic work or published in languages other than English were excluded. RESULTS AND CONCLUSIONS The review identifies key challenges related to the efficiency and variability of organoid generation and discusses ongoing efforts to enhance their predictive capabilities in drug screening and personalized medicine. Recent studies utilizing patient-derived organoid models for natural compound screening are highlighted, demonstrating the potential of these models in developing new classes of anticancer agents. The integration of natural products with patient-derived organoid models presents a promising approach for discovering novel anticancer compounds and elucidating their mechanisms of action.
Collapse
Affiliation(s)
- Rong Cong
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Can Lu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xinying Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yaqin Wang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Shusen Sun
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, USA
| |
Collapse
|
24
|
Nguyen TD, Winek MA, Rao MK, Dhyani SP, Lee MY. Nuclear envelope components in vascular mechanotransduction: emerging roles in vascular health and disease. Nucleus 2025; 16:2453752. [PMID: 39827403 DOI: 10.1080/19491034.2025.2453752] [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: 10/08/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
The vascular network, uniquely sensitive to mechanical changes, translates biophysical forces into biochemical signals for vessel function. This process relies on the cell's architectural integrity, enabling uniform responses to physical stimuli. Recently, the nuclear envelope (NE) has emerged as a key regulator of vascular cell function. Studies implicate nucleoskeletal elements (e.g. nuclear lamina) and the linker of nucleoskeleton and cytoskeleton (LINC) complex in force transmission, emphasizing nucleo-cytoskeletal communication in mechanotransduction. The nuclear pore complex (NPC) and its component proteins (i.e. nucleoporins) also play roles in cardiovascular disease (CVD) progression. We herein summarize evidence on the roles of nuclear lamina proteins, LINC complex members, and nucleoporins in endothelial and vascular cell mechanotransduction. Numerous studies attribute NE components in cytoskeletal-related cellular behaviors to insinuate dysregulation of nucleocytoskeletal feedback and nucleocytoplasmic transport as a mechanism of endothelial and vascular dysfunction, and hence implications for aging and vascular pathophysiology.
Collapse
Affiliation(s)
- Tung D Nguyen
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
- The Center for Cardiovascular Research, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Michael A Winek
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Mihir K Rao
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Shaiva P Dhyani
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Monica Y Lee
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
- The Center for Cardiovascular Research, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| |
Collapse
|
25
|
Mutsuda K, Nishii Y, Toyoshima T, Fukushima H, Motose H, Takahashi T. Specific enhancement of the translation of thermospermine-responsive uORF-containing mRNAs by ribosomal mutations in Arabidopsis thaliana. PLANT SIGNALING & BEHAVIOR 2025; 20:2480231. [PMID: 40088139 PMCID: PMC11913374 DOI: 10.1080/15592324.2025.2480231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/15/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Auxin-induced xylem formation in angiosperms is negatively regulated by thermospermine, whose biosynthesis is also induced by auxin. In Arabidopsis thaliana, loss-of-function mutants of ACL5, which encodes thermospermine synthase, exhibit a dwarf phenotype accompanied by excessive xylem formation. Studies of suppressor mutants that recover from the acl5 dwarf phenotype suggest that thermospermine alleviates the inhibitory effect of an upstream open-reading frame (uORF) on the main ORF translation of SAC51 mRNA. Many suppressor mutations for acl5 have been mapped to the uORF conserved in the SAC51 family or to ribosomal protein genes, such as RPL10A, RPL4A, and RACK1A. In this study, we identified newly isolated acl5 suppressors, sac501, sac504, and sac506, which are additional alleles of RPL10A and the uORFs of SAC51 family members, SACL1 and SACL3, respectively. To investigate whether acl5-suppressor alleles of ribosomal genes broadly affect translation of uORF-containing mRNAs, we examined GUS activity in several 5'-GUS fusion constructs. Our results showed that these alleles enhanced GUS activity in SAC51 and SACL3 5'-fusion constructs but had no effect on other 5'-fusion constructs unrelated to thermospermine response. This suggests that these ribosomal proteins are specifically involved in the thermospermine-mediated regulation of mRNA translation.
Collapse
Affiliation(s)
- Koki Mutsuda
- Graduate School of Environmental, Life, Natural Science and Technology, Okayama University, Okayama, Japan
| | - Yuichi Nishii
- Graduate School of Environmental, Life, Natural Science and Technology, Okayama University, Okayama, Japan
| | - Tomohiko Toyoshima
- Graduate School of Environmental, Life, Natural Science and Technology, Okayama University, Okayama, Japan
| | - Hiroko Fukushima
- Graduate School of Environmental, Life, Natural Science and Technology, Okayama University, Okayama, Japan
| | - Hiroyasu Motose
- Graduate School of Environmental, Life, Natural Science and Technology, Okayama University, Okayama, Japan
| | - Taku Takahashi
- Graduate School of Environmental, Life, Natural Science and Technology, Okayama University, Okayama, Japan
| |
Collapse
|
26
|
Deshpande G, Das S, Roy AE, Ratnaparkhi GS. A face-off between Smaug and Caspar modulates primordial germ cell count and identity in Drosophila embryos. Fly (Austin) 2025; 19:2438473. [PMID: 39718186 DOI: 10.1080/19336934.2024.2438473] [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: 09/03/2024] [Revised: 11/26/2024] [Accepted: 12/01/2024] [Indexed: 12/25/2024] Open
Abstract
Proper formation and specification of Primordial Germ Cells (PGCs) is of special significance as they gradually transform into Germline Stem Cells (GSCs) that are ultimately responsible for generating the gametes. Intriguingly, not only the PGCs constitute the only immortal cell type but several specific determinants also underlying PGC specification such as Vasa, Nanos and Germ-cell-less are conserved through evolution. In Drosophila melanogaster, PGC formation and specification depends on two independent factors, the maternally deposited specialized cytoplasm (or germ plasm) enriched in germline determinants, and the mechanisms that execute the even partitioning of these determinants between the daughter cells. Prior work has shown that Oskar protein is necessary and sufficient to assemble the functional germ plasm, whereas centrosomes associated with the nuclei that invade the germ plasm are responsible for its equitable distribution. Our recent data suggests that Caspar, the Drosophila orthologue of human Fas-associated factor-1 (FAF1) is a novel regulator that modulates both mechanisms that underlie the determination of PGC fate. Consistently, early blastoderm embryos derived from females compromised for caspar display reduced levels of Oskar and defective centrosomes.
Collapse
Affiliation(s)
- Girish Deshpande
- Department of Biology, Indian Institute of Science Education & Research, Pune, India
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Subhradip Das
- Department of Biology, Indian Institute of Science Education & Research, Pune, India
| | - Adheena Elsa Roy
- Department of Biology, Indian Institute of Science Education & Research, Pune, India
| | - Girish S Ratnaparkhi
- Department of Biology, Indian Institute of Science Education & Research, Pune, India
| |
Collapse
|
27
|
Forte G, Boteva L, Gilbert N, Cook PR, Marenduzzo D. Bridging-mediated compaction of mitotic chromosomes. Nucleus 2025; 16:2497765. [PMID: 40340634 PMCID: PMC12068332 DOI: 10.1080/19491034.2025.2497765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/27/2025] [Accepted: 04/21/2025] [Indexed: 05/10/2025] Open
Abstract
Within living cells, chromosome shapes undergo a striking morphological transition, from loose and uncondensed fibers during interphase to compacted and cylindrical structures during mitosis. ATP driven loop extrusion performed by a specialized protein complex, condensin, has recently emerged as a key driver of this transition. However, while this mechanism can successfully recapitulate the compaction of chromatids during the early stages of mitosis, it cannot capture structures observed after prophase. Here we hypothesize that a condensin bridging activity plays an additional important role, and review evidence - obtained largely through molecular dynamics simulations - that, in combination with loop extrusion, it can generate compact metaphase cylinders. Additionally, the resulting model qualitatively explains the unusual elastic properties of mitotic chromosomes observed in micromanipulation experiments and provides insights into the role of condensins in the formation of abnormal chromosome structures associated with common fragile sites.
Collapse
Affiliation(s)
- Giada Forte
- SUPA, School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| | - Lora Boteva
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Nick Gilbert
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Peter R. Cook
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Davide Marenduzzo
- SUPA, School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
28
|
Huang Y, Wang Z. Therapeutic potential of SOX family transcription factors in osteoarthritis. Ann Med 2025; 57:2457520. [PMID: 39887675 PMCID: PMC11789227 DOI: 10.1080/07853890.2025.2457520] [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: 06/22/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND As the worldwide population ages, osteoarthritis has significantly increased. This musculoskeletal condition has become a pressing global health issue and thus, prevention and treatment of osteoarthritis have become the primary focus of domestic and international research. Scholarly investigations of the molecular mechanisms that are related to the occurrence and development of osteoarthritis have shed light on the pathological causes of this condition to a certain extent, providing a foundation for its prevention and treatment. However, further research is necessary to fully understand the critical role of the transcription factor SOX9 in chondrocyte differentiation and the development of osteoarthritis. As a result, there has been widespread interest in SOX transcription factors. While SOX9 has been utilized as a biomarker to indicate the occurrence and prognosis of osteoarthritis, investigations into other members of the SOX family and the development of targeted treatments around SOX9 are still required. PURPOSE This article considers the impact of the SOX protein on the development and inhibition of osteoarthritis and highlights the need for therapeutic approaches targeting SOX9, as supported by existing research. RESULTS SOX9 can contribute to the process of osteoarthritis through acetylation and ubiquitination modifications. The regulation of the WNT signalling pathway, Nrf2/ARE signalling pathway, NF-κB signalling pathway and SOX9 is implicated in the emergence of osteoarthritis. Non-coding RNA may play a role in the onset and progression of osteoarthritis by modulating various SOX family members, including SOX2, SOX4, SOX5, SOX6, SOX8, SOX9 and SOX11. CONCLUSION SOX9 has the capability of mitigating the onset and progression of osteoarthritis through means such as medication therapy, stem cell therapy, recombinant adeno-associated virus (rAAV) vector therapy, physical therapy and other approaches.
Collapse
Affiliation(s)
- Yue Huang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Zhuo Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
29
|
Xie Y, Mi X, Xing Y, Dai Z, Pu Q. Past, present, and future of exosomes research in cancer: A bibliometric and visualization analysis. Hum Vaccin Immunother 2025; 21:2488551. [PMID: 40207548 PMCID: PMC11988232 DOI: 10.1080/21645515.2025.2488551] [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: 01/21/2025] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer seriously threatens the lives and health of people worldwide, and exosomes seem to play an important role in managing cancer effectively, which has attracted extensive attention from researchers in recent years. This study aimed to scientifically visualize exosomes research in cancer (ERC) through bibliometric analysis, reviewing the past, summarizing the present, and predicting the future, with a view to providing valuable insights for scholars and policy makers. Researches search and data collection from Web of Science Core Collection and clinical trial.gov. Calculations and visualizations were performed using Microsoft Excel, VOSviewer, Bibliometrix R-package, and CiteSpace. As of December 1, 2024, and March 8, 2025, we identified 8,001 ERC-related publications and 107 ERC-related clinical trials, with an increasing trend in annual publications. Our findings supported that China, Nanjing Medical University, and International Journal of Molecular Sciences were the most productive countries, institutions, and journals, respectively. Whiteside, Theresa L. had the most publications, while Théry, C was the most co-cited scholar. In addition, Cancer Research was the most co-cited journal. Spatial and temporal distribution of clinical trials was the same as for publications. High-frequency keywords were "extracellular vesicle," "microRNA" and "biomarker." Additional, "surface functionalization," "plant," "machine learning," "nanomaterials," "promotes metastasis," "engineered exosomes," and "macrophage-derived exosomes" were promising research topics. Our study comprehensively and visually summarized the structure, hotspots, and evolutionary trends of ERC. It would inspire subsequent studies from a macroscopic perspective and provide a basis for rational allocation of resources and identification of collaborations among researchers.
Collapse
Affiliation(s)
- Yafei Xie
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Xingqi Mi
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yikai Xing
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zhangyi Dai
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Qiang Pu
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
30
|
Pham-Bui HA, Lee M. Germ granule-mediated mRNA storage and translational control. RNA Biol 2025; 22:1-11. [PMID: 39895378 PMCID: PMC11810088 DOI: 10.1080/15476286.2025.2462276] [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: 10/05/2024] [Revised: 12/24/2024] [Accepted: 01/28/2025] [Indexed: 02/04/2025] Open
Abstract
Germ cells depend on specialized post-transcriptional regulation for proper development and function, much of which is mediated by dynamic RNA granules. These membrane-less organelles form through the condensation of RNA and proteins, governed by multivalent biomolecular interactions. RNA granules compartmentalize cellular components, selectively enriching specific factors and modulating biochemical reactions. Over recent decades, various types of RNA granules have been identified in germ cells across species, with extensive studies uncovering their molecular roles and developmental significance. This review explores the mRNA regulatory mechanisms mediated by RNA granules in germ cells. We discuss the distinct spatial organization of specific granule components and the variations in material states of germ granules, which contribute to the regulation of mRNA storage and translation. Additionally, we highlight emerging research on how changes in these material states, during developmental stages, reflect the dynamic nature of germ granules and their critical role in development.
Collapse
Affiliation(s)
- Hoang-Anh Pham-Bui
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan-si, Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si, Korea
| | - Mihye Lee
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan-si, Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si, Korea
| |
Collapse
|
31
|
Wu X, Xu H, Xia E, Gao L, Hou Y, Sun L, Zhang H, Cheng Y. Histone modifications in the regulation of erythropoiesis. Ann Med 2025; 57:2490824. [PMID: 40214280 PMCID: PMC11995772 DOI: 10.1080/07853890.2025.2490824] [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: 11/16/2024] [Revised: 03/13/2025] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
INTRODUCTION The pathogenesis of anemia and other erythroid dysphasia are mains poorly understood, primarily due to limited knowledge about the differentiation processes and regulatory mechanisms governing erythropoiesis. Erythropoiesis is a highly complex and precise biological process, that can be categorized into three distinct stages: early erythropoiesis, terminal erythroid differentiation, and reticulocyte maturation, and this complex process is tightly controlled by multiple regulatory factors. Emerging evidence highlights the crucial role of epigenetic modifications, particularly histone modifications, in regulating erythropoiesis. Methylation and acetylation are two common modification forms that affect genome accessibility by altering the state of chromatin, thereby regulating gene expression during erythropoiesis. DISCUSSION This review systematically examines the roles of histone methylation and acetylation, along with their respective regulatory enzymes, in regulating the development and differentiation of hematopoietic stem/progenitor cells (HSPCs) and erythroid progenitors. Furthermore, we discuss the involvement of these histone modifications in erythroid-specific developmental processes, including hemoglobin switching, chromatin condensation, and enucleation.Conclusions This review summarizes the current understanding of the role of histone modifications in erythropoiesis based on existing research, as a foundation for further research the mechanisms of epigenetic regulatory in erythropoiesis.
Collapse
Affiliation(s)
- Xiuyun Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Hongdi Xu
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Erxi Xia
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Linru Gao
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Yan Hou
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Lei Sun
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Hengchao Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Ying Cheng
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
32
|
Qing L, Qian X, Zhu H, Wang J, Sun J, Jin Z, Tang X, Zhao Y, Wang G, Zhao J, Chen W, Tian P. Maternal-infant probiotic transmission mitigates early-life stress-induced autism in mice. Gut Microbes 2025; 17:2456584. [PMID: 39931863 PMCID: PMC11817528 DOI: 10.1080/19490976.2025.2456584] [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: 11/09/2024] [Revised: 12/14/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025] Open
Abstract
Autism, a disorder influenced by both genetic and environmental factors, presents significant challenges for prevention and treatment. While maternal-infant gut microbiota has been a focus in autism research, preventive strategies targeting maternal gut microbiota remain underexplored. This study demonstrates that prenatal probiotic intake can effectively prevent maternal separation-induced autistic-like behaviors in offspring without altering the embryonic neurodevelopment in mice. Using specific PCR primers and cross-fostering experiments, we traced the vertical transmission of probiotics, primarily via fecal/vaginal contamination. Early probiotic colonization conferred resilience against stress-induced gut pathogenic microbes and Th17-mediated peripheral inflammation while significantly inhibiting hypermyelination and neuroinflammation linked to systemic inflammation. Microbial metabolites like tyrosol and xanthurenic acid alleviated neuroinflammation and hypermyelination in vitro, though the causal relationship among neuroinflammation, hypermyelination, and autism in vivo requires further validation. These findings underscore the importance of the maternal-infant microbiota transmission window in autism prevention and highlight the clinical potential of prenatal probiotic interventions.
Collapse
Affiliation(s)
- Li Qing
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xin Qian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Huiyue Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jingyu Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jingge Sun
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Zhiying Jin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xinyu Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Yingqi Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, P. R. China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Peijun Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| |
Collapse
|
33
|
Zhang H, Xu M. YTHDF1 activates FBW7 transcription by regulating m 6A-dependent FOXO1 to facilitate inflammatory response in ulcerative colitis-like model. Autoimmunity 2025; 58:2491717. [PMID: 40384634 DOI: 10.1080/08916934.2025.2491717] [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: 05/23/2024] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 05/20/2025]
Abstract
The prevalence of inflammatory bowel disease (IBD) has increased recently and lacks curative treatments. The involvement of the N6-methyladenosine (m6A) reader YTH N6-methyladenosine RNA binding protein 1 (YTHDF1) in ulcerative colitis (UC)-like model was studied in this study. DSS was employed to induce the UC-like condition both in vitro and in vivo. TNF-α, IL-1β, IL-6, and IL-8 secretion levels were analyzed by ELISA assay. Cell vitality was determined by CCK8 assay. FOXO1 mRNA m6A level was examined using methylated RNA binding protein immunoprecipitation (Me-RIP) assay. The interactions between YTHDF1 and FOXO1 were analyzed by RIP assay. ChIP and dual luciferase reporter assays were used to explore the relationship between FOXO1 and FBW7. YTHDF1, FOXO1, and FBW7 were overexpressed in DSS-induced colon epithelial cells. YTHDF1 downregulation alleviated DSS-induced inflammation and NF-κB signal activation in colon epithelial cells. Mechanically, YTHDF1 increased FOXO1 mRNA stability in an m6A manner. YTHDF1 overexpression prevented the inhibition of FOXO1 knockdown on DSS-induced inflammation in colon epithelial cells. In addition, FOXO1 transcriptionally activated FBW7. Moreover, FOXO1 upregulation abolished the inhibitory effect of FBW7 knockdown on DSS-induced inflammation in colon epithelial cells. Animal experiments also showed that YTHDF1 deletion alleviated inflammatory response in UC-like mice. YTHDF1 promoted inflammatory response in the UC-like model by transcriptionally activating FBW7 through regulating m6A-dependent FOXO1.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Gerontology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Meili Xu
- Department of Gerontology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| |
Collapse
|
34
|
Meza-Torres J, Tinevez JY, Crouzols A, Mary H, Kim M, Hunault L, Chamorro-Rodriguez S, Lejal E, Altamirano-Silva P, Groussard D, Gobaa S, Peltier J, Chassaing B, Dupuy B. Clostridioides difficile binary toxin CDT induces biofilm-like persisting microcolonies. Gut Microbes 2025; 17:2444411. [PMID: 39719371 DOI: 10.1080/19490976.2024.2444411] [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: 07/19/2024] [Revised: 11/07/2024] [Accepted: 12/09/2024] [Indexed: 12/26/2024] Open
Abstract
Clinical symptoms of Clostridioides difficile infection (CDI) range from diarrhea to pseudomembranous colitis. A major challenge in managing CDI is the high rate of relapse. Several studies correlate the production of CDT binary toxin by clinical strains of C. difficile with higher relapse rates. Although the mechanism of action of CDT on host cells is known, its exact contribution to CDI is still unclear. To understand the physiological role of CDT during CDI, we established two hypoxic relevant intestinal models, Transwell and Microfluidic Intestine-on-Chip systems. Both were challenged with the epidemic strain UK1 CDT+ and its isogenic CDT- mutant. We report that CDT induces mucin-associated microcolonies that increase C. difficile colonization and display biofilm-like properties by enhancing C. difficile resistance to vancomycin. Importantly, biofilm-like microcolonies were also observed in the cecum and colon of infected mice. Hence, our study shows that CDT induces biofilm-like microcolonies, increasing C. difficile persistence and risk of relapse.
Collapse
Affiliation(s)
- Jazmin Meza-Torres
- Pathogenesis of Bacterial Anaerobes, Department of Microbiology, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| | - Jean-Yves Tinevez
- Image Analysis Hub, Department of Cell Biology and Infection, Institut Pasteur, Université Paris Cité, Paris, France
| | - Aline Crouzols
- Pathogenesis of Bacterial Anaerobes, Department of Microbiology, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| | - Héloïse Mary
- Biomaterials and Microfluidics Core Facility, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Minhee Kim
- Biomaterials and Microfluidics Core Facility, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Lise Hunault
- Antibodies in Therapy and Pathology, Department of Immunology, Institut Pasteur, Paris, France
| | - Susan Chamorro-Rodriguez
- Pathogenesis of Bacterial Anaerobes, Department of Microbiology, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| | - Emilie Lejal
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, France
| | - Pamela Altamirano-Silva
- Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, San José, Costa Rica
| | | | - Samy Gobaa
- Biomaterials and Microfluidics Core Facility, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Johann Peltier
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, France
| | - Benoit Chassaing
- Microbiome-Host Interactions, Department of Microbiology, Institut Pasteur, Université Paris Cité, INSERM U1306, Paris, France
- Mucosal Microbiota in Chronic Inflammatory Diseases, INSERM U1016, CNRS UMR 8104, Université Paris Cité, Paris, France
| | - Bruno Dupuy
- Pathogenesis of Bacterial Anaerobes, Department of Microbiology, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| |
Collapse
|
35
|
Hartasánchez DA, Dumond M, Dubrulle N, Monéger F, Boudaoud A. Highly expressed cell wall genes contribute to robustness of sepal size. PLANT SIGNALING & BEHAVIOR 2025; 20:2446858. [PMID: 39739543 DOI: 10.1080/15592324.2024.2446858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/02/2025]
Abstract
Reproducibility in organ size and shape is a fascinating trait of living organisms. The mechanisms underlying such robustness remain, however, to be elucidated. Taking the sepal of Arabidopsis as a model, we investigated whether variability of gene expression plays a role in variation of organ size and shape. Previous work from our team identified cell-wall related genes as being enriched among the genes whose expression is highly variable. We then hypothesized that the variation of measured morphological parameters in cell-wall related single knockout mutants could be correlated with the variation in gene expression of the corresponding gene (the knocked-out gene) in wild-type plants. We analyzed sepal size and shape from 16 cell-wall mutants and found that sepal size variability correlates positively, not with gene expression variation, but with mean gene expression of the corresponding gene in wild type. These findings support a contribution of cell-wall related genes to the robustness of sepal size.
Collapse
Affiliation(s)
- Diego A Hartasánchez
- Laboratoire Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, CNRS, INRAE, UCBL, Lyon, France
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Mathilde Dumond
- Laboratoire Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, CNRS, INRAE, UCBL, Lyon, France
| | - Nelly Dubrulle
- Laboratoire Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, CNRS, INRAE, UCBL, Lyon, France
| | - Françoise Monéger
- Laboratoire Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, CNRS, INRAE, UCBL, Lyon, France
| | - Arezki Boudaoud
- Laboratoire Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, CNRS, INRAE, UCBL, Lyon, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau Cedex, France
| |
Collapse
|
36
|
Monreal Contreras HA, Arthikala MK, Lara M, Nanjareddy K. Target of Rapamycin is involved in root hair development in Phaseolus vulgaris. PLANT SIGNALING & BEHAVIOR 2025; 20:2507736. [PMID: 40390329 PMCID: PMC12101582 DOI: 10.1080/15592324.2025.2507736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2025] [Revised: 05/11/2025] [Accepted: 05/13/2025] [Indexed: 05/21/2025]
Abstract
Root hairs are essential for nutrient acquisition and rhizosphere interactions in vascular plants. While the Target of Rapamycin (TOR) kinase is a well established regulator of growth and metabolism, its role in root hair development in Phaseolus vulgaris remains underexplored. In this study, we investigated the role of TOR in root hair morphogenesis using RNA interference (RNAi)-mediated downregulation of PvTOR and transcriptomic profiling. Microscopic examination of PvTOR-RNAi roots confirmed significant reductions in root hair length and density. Transcriptomic analysis revealed differential expression of 148 P. vulgaris homologs of Arabidopsis thaliana root hair-related genes, with 63 genes downregulated and 85 upregulated. Gene Ontology enrichment analysis indicated that these differentially expressed genes (DEGs) were primarily involved in cellular development, cell differentiation, and redox regulation. Upregulation of phosphoinositide metabolism genes, ROS generators, and cell wall-related extensins suggests compensatory tip growth responses under TOR suppression. On the otherhand, repression of key auxin signaling genes and cell wall-loosening proteins such as EXPA1 and ENDOGLUCANASE5 indicates a shift away from elongation processes. Protein - protein interaction network analysis highlighted phosphoinositide and ROP GTPase signaling hubs as major pathways affected by TOR inhibition, suggesting that TOR indirectly modulates cell polarity and membrane dynamics essential for root hair development. These findings provide further evidence of TOR as a central integrator of hormonal, metabolic, and structural cues during root hair formation.
Collapse
Affiliation(s)
- Hugo Alberto Monreal Contreras
- Ciencias Agrogenómicas, Escuela Nacional de Estudios Superiores Unidad León-Universidad Nacional Autónoma de México (UNAM), Guanajuato, México
| | - Manoj-Kumar Arthikala
- Ciencias Agrogenómicas, Escuela Nacional de Estudios Superiores Unidad León-Universidad Nacional Autónoma de México (UNAM), Guanajuato, México
| | - Miguel Lara
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, México
| | - Kalpana Nanjareddy
- Ciencias Agrogenómicas, Escuela Nacional de Estudios Superiores Unidad León-Universidad Nacional Autónoma de México (UNAM), Guanajuato, México
| |
Collapse
|
37
|
He X, Zhu J, Gong X, Zhang D, Li Y, Zhang X, Zhao X, Zhou C. Advances in deciphering the mechanisms of salt tolerance in Maize. PLANT SIGNALING & BEHAVIOR 2025; 20:2479513. [PMID: 40098499 PMCID: PMC11959903 DOI: 10.1080/15592324.2025.2479513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/04/2025] [Accepted: 03/08/2025] [Indexed: 03/19/2025]
Abstract
Maize (Zea mays L.) is a vital crop worldwide, serving as a cornerstone for food security, livestock feed, and biofuel production. However, its cultivation is increasingly jeopardized by environmental challenges, notably soil salinization, which severely constrains growth, yield, and quality. To combat salinity stress, maize employs an array of adaptive mechanisms, including enhanced antioxidant enzyme activity and modulated plant hormone levels, which work synergistically to maintain reactive oxygen species (ROS) balance and ion homeostasis. This review explores the intricate interactions among ROS, antioxidant systems, plant hormones, and ion regulation in maize under salt stress, providing a comprehensive understanding of the physiological and molecular basis of its tolerance. By elucidating these mechanisms, this study contributes to the development of salt-tolerant maize varieties and informs innovative strategies to sustain agricultural productivity under adverse environmental conditions, offering significant theoretical insights into plant stress biology and practical solutions for achieving sustainable agriculture amidst global climate challenges.
Collapse
Affiliation(s)
- Xiaofei He
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
| | - Junke Zhu
- School of Agricultural Engineering & Food Science, Shandong University of Technology, Zibo, Shandong, China
- College of Life Sciences, Qilu Normal University, Jinan, Shandong, China
| | - Xuehua Gong
- Hebei Province Carbon-Based Heavy Metal Soil Pollution Remediation Technology Innovation Center, Tangshan, Hebei, China
| | - Dongqing Zhang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
| | - Yuan Li
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
| | - Xiansheng Zhang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
| | - Xiangyu Zhao
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
| | - Chao Zhou
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong, China
| |
Collapse
|
38
|
Sun Y, Wang Q, Jiang Y, He J, Jia D, Luo M, Shen W, Wang Q, Qi Y, Lin Y, Zhang Y, Wang L, Wang L, Chen S, Fan L. Lactobacillus intestinalis facilitates tumor-derived CCL5 to recruit dendritic cell and suppress colorectal tumorigenesis. Gut Microbes 2025; 17:2449111. [PMID: 39773173 PMCID: PMC11730368 DOI: 10.1080/19490976.2024.2449111] [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: 02/08/2024] [Revised: 11/11/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Gut microbes play a crucial role in regulating the tumor microenvironment (TME) of colorectal cancer (CRC). Nevertheless, the deep mechanism between the microbiota-TME interaction has not been well explored. In this study, we for the first time discovered that Lactobacillus intestinalis (L. intestinalis) effectively suppressed tumor growth both in the AOM/DSS-induced CRC model and the ApcMin/+ spontaneous adenoma model. Our investigation revealed that L. intestinalis increased the infiltration of immune cells, particularly dendritic cells (DC), in the TME. Mechanically, the tumor-derived CCL5 induced by L. intestinalis recruited DC chemotaxis through the NOD1/NF-κB signaling pathway. In clinical samples and datasets, we found positive correlation between L. intestinalis, CCL5 level, and the DC-related genes. Our study provided a new strategy for microbial intervention for CRC and deepened the understanding of the interaction between tumor cells and the immune microenvironment modulated by gut microbes.
Collapse
Affiliation(s)
- Yong Sun
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Qiwen Wang
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yao Jiang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jiamin He
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Dingjiacheng Jia
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Man Luo
- Department of Nutrition, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Wentao Shen
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Qingyi Wang
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yadong Qi
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yifeng Lin
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Ying Zhang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Lan Wang
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Liangjing Wang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
- Prevention and Treatment Research Center of Senescent Disease, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Shujie Chen
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Prevention and Treatment Research Center of Senescent Disease, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Lina Fan
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
39
|
Szabó A, Borkúti P, Kovács Z, Kristó I, Vilmos P. Recent advances in nuclear actin research. Nucleus 2025; 16:2498643. [PMID: 40320716 PMCID: PMC12054378 DOI: 10.1080/19491034.2025.2498643] [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: 02/14/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/08/2025] Open
Abstract
Actin was first observed in the nucleus more than sixty years ago but research on nuclear actin did not receive significant attention for the next forty years. It only started to accelerate around the year 2000, when the first convincing experimental data emerged indicating that actin participates in essential nuclear processes. Today, we know that actin is involved in transcription, replication, DNA repair, chromatin remodeling, and participates in the determination of nuclear shape and size. In this paper we review the results of the last five years of increasingly intensive research on nuclear actin, because on one hand, the field has expanded with several new directions during this time, and on the other hand, the enrichment of our picture of nuclear actin will certainly provide a more solid foundation and new impetus for its future investigation.
Collapse
Affiliation(s)
- Anikó Szabó
- HUN-REN Biological Research Centre, Institute of Genetics, Szeged, Hungary
| | - Péter Borkúti
- HUN-REN Biological Research Centre, Institute of Genetics, Szeged, Hungary
| | - Zoltán Kovács
- HUN-REN Biological Research Centre, Institute of Genetics, Szeged, Hungary
| | - Ildikó Kristó
- HUN-REN Biological Research Centre, Institute of Genetics, Szeged, Hungary
| | - Péter Vilmos
- HUN-REN Biological Research Centre, Institute of Genetics, Szeged, Hungary
| |
Collapse
|
40
|
Lee JS, Kao DJ, Worledge CS, Villamaria ZF, Wang RX, Welch NM, Kostelecky RE, Colgan SP. E. coli genetically modified for purine nucleobase release promotes butyrate generation and colonic wound healing during DSS insult. Gut Microbes 2025; 17:2490211. [PMID: 40247632 PMCID: PMC12013446 DOI: 10.1080/19490976.2025.2490211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 12/27/2024] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
The gut microbiota transforms energy stored as undigestible carbohydrates into a remarkable number of metabolites that fuel intestinal bacterial communities and the host tissue. Colonic epithelial cells at the microbiota-host interface depend upon such microbiota-derived metabolites (MDMs) to satisfy their energy requisite. Microbial dysbiosis eliciting MDM loss contributes to barrier dysfunction and mucosal disease. Recent work has identified a role for microbiota-sourced purines (MSPs), notably hypoxanthine, as an MDM salvaged by the colonic epithelium for nucleotide biogenesis and energy balance. Here, we investigated the role of MSPs in mice during disease-modeled colonic energetic stress using a strain of E. coli genetically modified for enhanced purine nucleobase release (E. coli Mutant). E. coli Mutant colonization protected against DSS-induced tissue damage and permeability while promoting proliferation for wound healing. Metabolite and metagenomic analyses suggested a colonic butyrate-purine nucleobase metabolic axis, wherein the E. coli Mutant provided purine substrate for Clostridia butyrate production and host purine salvage, altogether supplying the host substrate for efficient nucleotide biogenesis and energy balance.
Collapse
Affiliation(s)
- J. Scott Lee
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Daniel J. Kao
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Corey S. Worledge
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Zachary F. Villamaria
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Ruth X. Wang
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Nichole M. Welch
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
- Department of Medicine, Rocky Mountain Veterans Association, Aurora, CO, USA
| | - Rachael E. Kostelecky
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Sean P. Colgan
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
- Department of Medicine, Rocky Mountain Veterans Association, Aurora, CO, USA
| |
Collapse
|
41
|
Chagraoui A, Thibaut F, De Deurwaerdère P. 5-HT6 receptors: Contemporary views on their neurobiological and pharmacological relevance in neuropsychiatric disorders. DIALOGUES IN CLINICAL NEUROSCIENCE 2025; 27:112-128. [PMID: 40347153 PMCID: PMC12068339 DOI: 10.1080/19585969.2025.2502028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 04/02/2025] [Accepted: 04/30/2025] [Indexed: 05/12/2025]
Abstract
Despite the relatively limited number of serotonergic neurons in humans, serotonin plays a key role in neurophysiological functions, including sleep, pain perception, learning, memory, cognition, emotion, reward, and mood regulation. Altered serotonergic neurotransmission is linked to conditions such as anxiety, depression, anorexia, migraine, insomnia, schizophrenia, Alzheimer's disease (AD), and cognitive impairments. The 5-HT6 receptor (5-HT6R), mainly found in brain regions involved in cognition, is a promising therapeutic target for cognitive deficits in neuropsychiatric disorders, particularly AD and schizophrenia. Preclinical studies have shown that 5-HT6R antagonists improve cognitive function. 5-HT6R interacts dynamically with an extensive intracellular protein network, regulating the localisation, trafficking, and signalling of these proteins. Proteomic and genetic studies have revealed interactions with mTOR kinase and neurofibromin, both of which are crucial for synaptic plasticity, learning, and memory. Fyn kinase is also associated with 5-HT6Rs, reinforcing receptor expression and G-protein coupling. Notably, the G protein-regulated inducer of neurite outgrowth 1 (GPRIN1) interacts with 5-HT6Rs independently of agonists, enhancing receptor activity. This review highlights the clinical testing of 5-HT6R ligands as regulators of these complex signalling properties, underscoring their therapeutic potential in addressing cognitive impairments associated with neuropsychiatric disorders.
Collapse
Affiliation(s)
- Abdeslam Chagraoui
- Department of Medical Biochemistry, Rouen University Hospital, CHU de Rouen, France
- University of Rouen, Faculty of Medicine and Pharmacy, Inserm U1239, Neuroendocrine, Endocrine, and Germinal Differentiation and Communication (NorDiC), Mont-Saint-Aignan, France
| | - Florence Thibaut
- Department of Psychiatry and Addictive Disorders, University Hospital Cochin (site Tarnier) AP-HP, Paris, France
- INSERM U1266, Institute of Psychiatry and Neurosciences, University of Paris, Cité, Paris, France
| | - Philippe De Deurwaerdère
- Centre National de la Recherche Scientifique, Institut des Neurosciences Intégratives et Cognitives d’Aquitaine, UMR 5287, Bordeaux, France
| |
Collapse
|
42
|
Chen R, Chai X, Zhang Y, Zhou T, Xia Y, Jiang X, Lv B, Zhang J, Zhou L, Tian X, Wang R, Mao L, Zhao F, Zhang H, Hu J, Qiu J, Zou Z, Chen C. Novel role of FTO in regulation of gut-brain communication via Desulfovibrio fairfieldensis-produced hydrogen sulfide under arsenic exposure. Gut Microbes 2025; 17:2438471. [PMID: 39852343 PMCID: PMC11776478 DOI: 10.1080/19490976.2024.2438471] [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: 12/21/2023] [Revised: 10/26/2024] [Accepted: 11/26/2024] [Indexed: 01/26/2025] Open
Abstract
Fat mass and obesity-associated protein (FTO) is the key demethylase that reverses the abnormally altered N6-methyladenosine (m6A) modification in eukaryotic cells under environmental pollutants exposure. Arsenic is an environmental metalloid and can cause severe symptoms in human mainly through drinking water. However, there is no specific treatment for its toxic effects due to the uncovered mechanisms. We previously revealed that exposure to arsenic increased the level of m6A via down-regulation of FTO, which might serve as a potential target for intervention against arsenic-related disorders. In this study, our results demonstrated that chronic exposure to arsenic significantly disrupted the intestinal barrier and microenvironment. Also, this administration resulted in the enhancement of m6A modification and the reduction of FTO expression in the intestine. By using both CRISPR/Cas9-based FTO knock-in strategy and adeno-associated virus (AAV)-mediated overexpression of FTO in the intestine, we established for the first time that up-regulation of FTO remarkably ameliorated arsenic-induced disruption of intestinal barriers and altered microenvironment of mice. We also firstly identified a dominant gut microbial species, Desulfovibrio fairfieldensis, which was sharply reduced in arsenic-exposed mice, was able to proceed arsenic-induced neurobehavioral impairments by declining the levels of its major metabolite hydrogen sulfide. Administration of Desulfovibrio fairfieldensis could significantly alleviate the neurotoxicity of arsenic. Intriguingly, the beneficial effects of FTO against arsenic neurotoxicity possibly occurred through a novel gut-brain communication via Desulfovibrio fairfieldensis and its produced hydrogen sulfide. Collectively, these findings will provide new ideas for understanding the mechanisms of arsenic-induced toxic effects from a gut-brain communication perspective, and will assist the development of explicit intervention strategy via regulation of a new potential target FTO for prevention and treatment against arsenic-related both intestinal and neurological disorders.
Collapse
Affiliation(s)
- Ruonan Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xiaoqin Chai
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yunxiao Zhang
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Tianxiu Zhou
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yinyin Xia
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Bo Lv
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Lixiao Zhou
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ruonan Wang
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Lejiao Mao
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Feng Zhao
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Hongyang Zhang
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jun Hu
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Jingfu Qiu
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
43
|
Xue N, Zhao J, Yin J, Liu L, Yang Z, Zhai S, Bian X, Gao X. The Role of SUMO1 Modification of SOX9 in Cartilage Development Stimulated by Zinc Ions in Mice. Organogenesis 2025; 21:2460269. [PMID: 39905673 PMCID: PMC11801356 DOI: 10.1080/15476278.2025.2460269] [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: 07/18/2024] [Revised: 12/09/2024] [Accepted: 01/25/2025] [Indexed: 02/06/2025] Open
Abstract
Zinc ions play a pivotal role in facilitating the development of cartilage in mice. Nevertheless, the precise underlying mechanism remains elusive. Our investigation was centered on elucidating the impact of zinc deficiency on cartilage maturation by modulating SUMO1 and UBC9 at both the protein and mRNA levels. We administered a regimen inducing zinc deficiency to gravid mice from E0.5 until euthanasia. Subsequently, we subjected the embryos to scrutiny employing HE, Safranin O staining and IHC. Primary chondrocytes were isolated from fetal mouse femoral condyles and utilized for Western blot analysis to discern the expression profiles of SUMO1, SUMO2/3, UBC9, SOX9, MMP13, Collagen II, RUNX2, and aggrecan. Furthermore, ATDC5 murine chondrocytes were subjected to treatment with ZnCl2, followed by RT-PCR assessment to scrutinize the expression levels of MMP13, Collagen II, RUNX2, and aggrecan. Additionally, we conducted Co-IP assays on ZnCl2-treated ATDC5 cells to explore the interaction between SOX9 and SUMO1. Our investigation unveiled that zinc deficiency led to a reduction in cartilage development, as evidenced by the HE results in fetal murine femur. Moreover, diminished expression levels of SUMO1 and UBC9 were observed in the IHC and Western blot results. Furthermore, Western blot and Co-IP assays revealed an augmented interaction between SOX9 and SUMO1, which was potentiated by ZnCl2 treatment. Significantly, mutations at the SUMOylation site of SOX9 resulted in alterations in the expression patterns of crucial chondrogenesis factors. This research underscores how zinc ions promote cartilage development through the modification of SOX9 by SUMO1.
Collapse
Affiliation(s)
- Na Xue
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, China
- Emergency Medicine Department, Tianjin Fifth Central Hospital, Tianjin, China
| | - Jing Zhao
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Jing Yin
- Department of Pathology, Tianjin Fifth Central Hospital, Tianjin, China
| | - Liang Liu
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Zhong Yang
- Department of Orthopedics, Tianjin Fifth Central Hospital, Tianjin, China
| | - Shuchao Zhai
- Department of Orthopedics, Tianjin Fifth Central Hospital, Tianjin, China
| | - Xiyun Bian
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, China
| | - Xiang Gao
- Department of Orthopedics, Tianjin Fifth Central Hospital, Tianjin, China
| |
Collapse
|
44
|
Zeng Q, Xue L, Li W, Liang C, Zhou W, Xiong W, Dai X. Inhibition of 5-hydroxyindoleacetic acid to reduce neutrophil extracellular trap production improves lung condition in chronic obstructive pulmonary disease mice. Ann Med 2025; 57:2474734. [PMID: 40066951 PMCID: PMC11899248 DOI: 10.1080/07853890.2025.2474734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/09/2025] [Accepted: 01/31/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Neutrophil extracellular trap (NET) correlate with chronic obstructive pulmonary disease (COPD) severity. Platelets can promote NET generation. However, serotonin alone or serotonin-deficient platelets do not adequately promote NET production. The metabolism of serotonin to 5-hydroxyindoleacetic acid (5-HIAA) in platelets may be the key to this difference. OBJECTIVE The study aimed to determine whether 5-HIAA can influence NET production and thus play a role in COPD. METHODS After a 4-hour co-incubation with lipopolysaccharide (LPS) and 5-HIAA, NET and ROS levels in the culture medium were measured by ELISA, and NET production with aryl hydrocarbon receptor (AHR) expression in adherent cells were analyzed by immunofluorescence.A COPD model was established in C57BL/6 mice through smoke exposure combined with LPS tracheal administration, followed by selegiline or 5-HIAA treatment. Post-intervention, lung function tests and sample collection were performed. The levels of 5-HIAA, ROS, NET, IL-6, and AHR in the samples were quantified by ELISA, pathological changes were assessed by HE staining, and NET/AHR expression was detected by immunofluorescence. RESULTS 5-HIAA promoted NET production in vitro, and the nuclei of neutrophils secreting NET-like structures express AHR. In animal experiments, 5-HIAA levels were higher in both the plasma and lung tissues of COPD mice compared with normal mice. Inhibition of 5-HIAA in COPD mice down-regulated AHR expression, reduced reactive oxygen species and NET generation, elevated lung function indices (FEV0.1, FVC, PEF, and FEV0.1/FVC), decreased interleukin-6 levels, and improved lung tissue condition. CONCLUSION Inhibiting 5-HIAA reduces NET generation, thereby improving lung conditions in COPD mice, which is associated with the 5-HIAA/AHR pathway.
Collapse
Affiliation(s)
- Qiang Zeng
- Department of Geriatrics, First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Lei Xue
- Department of Geriatrics, First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Wu Li
- Department of Geriatrics, First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Cheng Liang
- Department of Geriatrics, First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Weijia Zhou
- Department of Geriatrics, First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Wei Xiong
- Department of Geriatrics, First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Xiaotian Dai
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
45
|
Xie X, Liu Y, Yang L, Zhang Z, Li H, Zhang W, Liu H, Wang H, Shao Z. Impaired LTB4-induced neutrophil chemotactic directionality in myelodysplastic neoplasms patients. Hematology 2025; 30:2483551. [PMID: 40170327 DOI: 10.1080/16078454.2025.2483551] [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: 08/25/2024] [Accepted: 03/19/2025] [Indexed: 04/03/2025] Open
Abstract
OBJECTIVES Myelodysplastic neoplasm (MDS) patients are at a high risk of infections, contributing significantly to morbidity and mortality. While neutrophil dysfunction is considered a primary factor, specific functional defects remain elusive. METHODS We conducted a comprehensive study involving 90 participants, including controls and de novo MDS patients. We utilized the TAXIScan-FL system to evaluate neutrophil chemotaxis towards leukotriene B4 (LTB4). The global reactive oxygen species (ROS) production by neutrophils were measured by chemiluminescence assay, neutrophil alkaline phosphatase (NAP) was evaluated by enzymatic staining. RESULTS MDS patients, irrespective of absolute neutrophil count (ANC) levels, exhibited elevated empirical antimicrobial therapy (EAT) rate compared to controls. Neutrophil migration towards LTB4 was notably impaired, demonstrating reduced velocity and directionality. Interestingly, MDS patients with high ANC still displayed poor directionality and slower migration. MDS patients also had compromised ROS and NAP activity. A noteworthy correlation was observed between EAT rate and chemotactic directionality parameters. CONCLUSION MDS patients face a heightened risk of infection, potentially attributed to impaired neutrophil chemotactic speed and directionality, alongside compromised ROS and NAP activity. Notably, chemotactic directionality emerged as a pivotal factor correlated with antimicrobial therapy. These insights hold significant clinical implications for managing infections in MDS patients, underscoring the importance of targeting specific neutrophil defects for more effective therapeutic strategies.
Collapse
Affiliation(s)
- Xinyan Xie
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Yumei Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Liyan Yang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Zhe Zhang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Hongzhao Li
- Tianjin Children's Hospital, Tianjin, People's Republic of China
| | - Wei Zhang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Hong Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Huaquan Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Zonghong Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| |
Collapse
|
46
|
Afrose D, Alfonso-Sánchez S, McClements L. Targeting oxidative stress in preeclampsia. Hypertens Pregnancy 2025; 44:2445556. [PMID: 39726411 DOI: 10.1080/10641955.2024.2445556] [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: 09/21/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Preeclampsia is a complex condition characterized by elevated blood pressure and organ damage involving kidneys or liver, resulting in significant morbidity and mortality for both the mother and the fetus. Increasing evidence suggests that oxidative stress, often caused by mitochondrial dysfunction within fetal trophoblast cells may play a major role in the development and progression of preeclampsia. Oxidative stress occurs as a result of an imbalance between the production of reactive oxygen species (ROS) and the capacity of antioxidant defenses, which can lead to placental cellular damage and endothelial cell dysfunction. Targeting oxidative stress appears to be a promising therapeutic approach that has the potential to improve both short- and long-term maternal and fetal outcomes, thus reducing the global burden of preeclampsia. The purpose of this review is to provide a comprehensive account of the mechanisms of oxidative stress in preeclampsia. Furthermore, it also examines potential interventions for reducing oxidative stress in preeclampsia, including natural antioxidant supplements, lifestyle modifications, mitochondrial targeting antioxidants, and pharmacological agents.A better understanding of the mechanism of action of proposed therapeutic strategies targeting oxidative stress is essential for the identification of companion biomarkers and personalized medicine approaches for the development of effective treatments of preeclampsia.
Collapse
Affiliation(s)
- Dinara Afrose
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Sofía Alfonso-Sánchez
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
47
|
Liu C, Wang P, Gao Y, Ma X, Su Y, Wei Y, Qiao R. CD39-Diannexin alleviates the platelet storage lesion by protecting platelets from activation, a new attempt from a traditional perspective. Platelets 2025; 36:2517108. [PMID: 40491155 DOI: 10.1080/09537104.2025.2517108] [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: 11/06/2024] [Revised: 05/05/2025] [Accepted: 06/02/2025] [Indexed: 06/11/2025]
Abstract
Due to platelet storage lesions (PSL), transfused platelets are unable to function properly in the prevention and treatment of bleeding in critically ill patients. It is a traditional assumption that PSL is closely related to platelet activation during storage because of the exposure of CD62P, phosphatidylserine (PS), etc. In this standpoint, activated platelets in vitro cannot be reactivated in vivo to exert their hemostatic function and exposed PS accelerates platelet clearance. Therefore, reducing platelet activation is helpful to alleviate PSL. Diannexin is the dimer of annexin that has a higher affinity for PS. CD39 is an ADP hydrolase produced by the vascular endothelium. As a result, we construct CD39-Diannexin (CD39-DA) fusion protein and hypothesize that CD39-DA can reduce platelet activation during storage to alleviate PSL. CD39-DA can bind to the exposed PS on the surface of stored platelets by immunofluorescence. Compared to the control groups, CD39-DA reserves part of stored platelets' aggregation function confirmed by platelet aggregation assay, induced by AA, ADP and collagen. Additionally, CD39-DA reduces lactic dehydrogenase (LDH) levels and CD62P-positive events after three-day storage. Interestingly, we preliminarily discover that CD39-DA may reduce stored platelets' apoptosis and increase aggregatory platelets after activation by thrombin, collagen and calcium, which is marked by GSAO. In conclusion, we confirm that CD39-DA can alleviate PSL by reducing platelet activation.
Collapse
Affiliation(s)
- Cheng Liu
- The Department of Laboratory Medicine, Peking University Third Hospital, Bei Jing, China
| | - Peng Wang
- Department of Transfusion, Peking University First Hospital, Bei Jing, China
| | - Yafei Gao
- The Department of Laboratory Medicine, Peking University Third Hospital, Bei Jing, China
| | - Xiaolong Ma
- The Department of Laboratory Medicine, Peking University Third Hospital, Bei Jing, China
| | - Yang Su
- The Department of Laboratory Medicine, Peking University Third Hospital, Bei Jing, China
| | - Yao Wei
- The Department of Laboratory Medicine, Peking University Third Hospital, Bei Jing, China
| | - Rui Qiao
- The Department of Laboratory Medicine, Peking University Third Hospital, Bei Jing, China
| |
Collapse
|
48
|
Kong M, Li J, Tong N. The role of peripheral blood microRNAs in the pathogenesis and treatment response of age-related macular degeneration. Future Sci OA 2025; 11:2482499. [PMID: 40183456 PMCID: PMC11980467 DOI: 10.1080/20565623.2025.2482499] [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: 11/02/2024] [Accepted: 02/12/2025] [Indexed: 04/05/2025] Open
Abstract
Age-related macular degeneration is a leading cause of vision loss in aging populations, driven by complex interactions between genetic, environmental, and molecular factors. MicroRNAs have emerged as crucial regulators of cellular processes such as oxidative stress, inflammation, and angiogenesis, all of which contribute to AMD pathogenesis. This narrative review aims to summarize the involvement of peripheral blood microRNAs in the pathogenesis of AMD, focusing on key pathways such as oxidative stress, inflammation, and angiogenesis. Additionally, it explores their potential as biomarkers for predicting treatment response, particularly to anti-VEGF therapies. The potential of miRNAs as noninvasive biomarkers for early diagnosis and personalized treatment strategies is also explored, highlighting future directions for research.
Collapse
Affiliation(s)
- Meng Kong
- Department of Ophthalmology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, China
- School of Medicine, Qingdao University, Qingdao, Shandong, China
| | - Jingwen Li
- Department of Ophthalmology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, China
| | - Nianting Tong
- Department of Ophthalmology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, China
- School of Medicine, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
49
|
Tumentemur G, Aygun EG, Yurtsever B, Cakirsoy D, Ovali E. Effect of amniotic fluid on hair follicle growth. J DERMATOL TREAT 2025; 36:2451389. [PMID: 39827901 DOI: 10.1080/09546634.2025.2451389] [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: 10/17/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
Purpose: Human amniotic fluid stem cells (hAFSCs) have shown significant regenerative potential in treating hair loss, wound healing, and tissue repair. This study aims to evaluate the effects of human amniotic fluid (hAF) on hair follicle (HF) regeneration and immune system modulation. Materials and Methods: The hAF used was pooled, acellular, and gamma-irradiated to standardize its contents and enhance its stability. Both irradiated (FAFI) and non-irradiated (FAF) hAF were assessed for their efficacy and safety in promoting hair growth and modulating immune responses in a rat model of hair loss. The study examined HF regeneration, transition to the anagen phase, and macrophage polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype. Results: Both FAF and FAFI treatments significantly increased HF density, with FAFI exhibiting enhanced effects. Histological analysis demonstrated improved HF regeneration, increased M2 macrophages, and reduced collagen fiber deposition in treated areas. Gamma irradiation likely improved the efficacy of FAFI by stabilizing active components and inhibiting protease activity. Conclusions: Irradiated hAF is a safe and effective therapeutic candidate for alopecia and HF growth disorders. These findings support further evaluation of hAF in clinical trials to validate its potential for hair regeneration therapies.
Collapse
Affiliation(s)
- Gamze Tumentemur
- Vocational School of Health Services, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Elif Ganime Aygun
- Department of Obstetrics and Gynecology, Acibadem Mehmet Ali Aydinlar University Atakent Hospital, Istanbul, Turkey
| | - Bulut Yurtsever
- Acibadem Labcell Cellular Therapy Laboratory, Istanbul, Turkey
| | - Didem Cakirsoy
- Acibadem Labcell Cellular Therapy Laboratory, Istanbul, Turkey
| | - Ercument Ovali
- Acibadem Labcell Cellular Therapy Laboratory, Istanbul, Turkey
| |
Collapse
|
50
|
Rayêe D, Meier UT, Eliscovich C, Cvekl A. Nucleolar ribosomal RNA synthesis continues in differentiating lens fiber cells until abrupt nuclear degradation required for ocular lens transparency. RNA Biol 2025; 22:1-16. [PMID: 40126102 PMCID: PMC11959900 DOI: 10.1080/15476286.2025.2483118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 02/20/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Cellular differentiation requires highly coordinated action of all three transcriptional systems to produce rRNAs, mRNAs and various 'short' and 'long' non-coding RNAs by RNA Polymerase I, II and III systems, respectively. RNA Polymerase I catalyzes transcription of about 400 copies of mammalian rDNA genes, generating 18S, 5.8S and 28S rRNA molecules. Lens fiber cell differentiation is a unique process to study transcriptional mechanisms of individual crystallin genes as their very high transcriptional outputs are directly comparable only to globin genes in erythrocytes. Importantly, both terminally differentiated lens fiber cells and mammalian erythrocytes degrade their nuclei through different mechanisms. In lens, the generation of the organelle-free zone (OFZ) includes the degradation of mitochondria, endoplasmic reticulum, Golgi apparatus and nuclei. Here, using RNA fluorescence in situ hybridization (FISH), we evaluated nascent rRNA transcription, located in the nucleoli, during the process of mouse lens fiber cell differentiation. Lens fiber cell nuclei undergo morphological changes including chromatin condensation prior to their denucleation. Remarkably, nascent rRNA transcription persists in all nuclei that are in direct proximity of the OFZ. Additionally, changes in both nuclei and nucleoli shape were evaluated via immunofluorescence detection of fibrillarin, nucleolin, UBF and other proteins. These studies demonstrate for the first time that highly condensed lens fiber cell nuclei have the capacity to support nascent rRNA transcription. Thus, we propose that 'late' production of rRNA molecules and consequently of ribosomes increases crystallin protein synthesis machinery within the mature lens fibers.
Collapse
Affiliation(s)
- Danielle Rayêe
- Departments of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - U. Thomas Meier
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Carolina Eliscovich
- Departments of Medicine (Hepatology) and Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aleš Cvekl
- Departments of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|