1
|
Hashemolhosseini S, Gessler L. Crosstalk among canonical Wnt and Hippo pathway members in skeletal muscle and at the neuromuscular junction. Neural Regen Res 2025; 20:2464-2479. [PMID: 39248171 PMCID: PMC11801303 DOI: 10.4103/nrr.nrr-d-24-00417] [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: 04/15/2024] [Revised: 07/04/2024] [Accepted: 08/05/2024] [Indexed: 09/10/2024] Open
Abstract
Skeletal muscles are essential for locomotion, posture, and metabolic regulation. To understand physiological processes, exercise adaptation, and muscle-related disorders, it is critical to understand the molecular pathways that underlie skeletal muscle function. The process of muscle contraction, orchestrated by a complex interplay of molecular events, is at the core of skeletal muscle function. Muscle contraction is initiated by an action potential and neuromuscular transmission requiring a neuromuscular junction. Within muscle fibers, calcium ions play a critical role in mediating the interaction between actin and myosin filaments that generate force. Regulation of calcium release from the sarcoplasmic reticulum plays a key role in excitation-contraction coupling. The development and growth of skeletal muscle are regulated by a network of molecular pathways collectively known as myogenesis. Myogenic regulators coordinate the differentiation of myoblasts into mature muscle fibers. Signaling pathways regulate muscle protein synthesis and hypertrophy in response to mechanical stimuli and nutrient availability. Several muscle-related diseases, including congenital myasthenic disorders, sarcopenia, muscular dystrophies, and metabolic myopathies, are underpinned by dysregulated molecular pathways in skeletal muscle. Therapeutic interventions aimed at preserving muscle mass and function, enhancing regeneration, and improving metabolic health hold promise by targeting specific molecular pathways. Other molecular signaling pathways in skeletal muscle include the canonical Wnt signaling pathway, a critical regulator of myogenesis, muscle regeneration, and metabolic function, and the Hippo signaling pathway. In recent years, more details have been uncovered about the role of these two pathways during myogenesis and in developing and adult skeletal muscle fibers, and at the neuromuscular junction. In fact, research in the last few years now suggests that these two signaling pathways are interconnected and that they jointly control physiological and pathophysiological processes in muscle fibers. In this review, we will summarize and discuss the data on these two pathways, focusing on their concerted action next to their contribution to skeletal muscle biology. However, an in-depth discussion of the non-canonical Wnt pathway, the fibro/adipogenic precursors, or the mechanosensory aspects of these pathways is not the focus of this review.
Collapse
Affiliation(s)
- Said Hashemolhosseini
- Institute of Biochemistry, Medical Faculty, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Lea Gessler
- Institute of Biochemistry, Medical Faculty, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
2
|
Lee S, Lee B, Kwon SH, Park J, Kim SH. MCC in the spotlight: Its dual role in signal regulation and oncogenesis. Cell Signal 2025; 131:111756. [PMID: 40118128 DOI: 10.1016/j.cellsig.2025.111756] [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: 01/24/2025] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
The mutated in colorectal cancer (MCC) gene is closely associated with the onset and progression of colorectal cancer. MCC plays a critical role in regulating the cell cycle and various signaling pathways and is recognized to inhibit cancer cell proliferation via the β-catenin signaling pathway. β-catenin is a key component of the WNT signaling pathway that influences cell growth, differentiation, survival, and migration, thereby positioning MCC as an important tumor suppressor. Notably, MCC has also been implicated in other cancer types, including lung, liver, and brain cancers. However, the precise mechanisms by which MCC functions in these malignancies remain inadequately understood. Comprehensive investigations into the interactions among MCC, various signaling pathways, and metabolic processes are essential for uncovering the molecular mechanisms of cancer and the pathological features characteristic of different cancer stages. This review presents the structural characteristics of MCC and its cell growth regulation mechanisms and functional roles within tissues, with the aims of enhancing our understanding of the role of MCC in cancer biology and highlighting potential therapeutic strategies targeting this gene.
Collapse
Affiliation(s)
- Soohyeon Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Beomwoo Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - So Hee Kwon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, South Korea.
| | - Jongsun Park
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Biomedical Research Institute, Chungnam National University Hospital, Daejeon 35015, Republic of Korea.
| | - Seon-Hwan Kim
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon 35015, Republic of Korea; Department of Neurosurgery, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, South Korea.
| |
Collapse
|
3
|
Chaudhary JK, Danga AK, Kumari A, Bhardwaj A, Rath PC. Role of stem cells in ageing and age-related diseases. Mech Ageing Dev 2025; 225:112069. [PMID: 40324541 DOI: 10.1016/j.mad.2025.112069] [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: 01/17/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Stem cell functions and ageing are deeply interconnected, continually influencing each other in multiple ways. Stem cells play a vital role in organ maintenance, regeneration, and homeostasis, all of which decline over time due to gradual reduction in their self-renewal, differentiation, and growth factor secretion potential. The functional decline is attributed to damaging extrinsic environmental factors and progressively worsening intrinsic genetic and biochemical processes. These ageing-associated deteriorative changes have been extensively documented, paving the way for the discovery of novel biomarkers of ageing for detection, diagnosis, and treatment of age-related diseases. Age-dependent changes in adult stem cells include numerical decline, loss of heterogeneity, and reduced self-renewal and differentiation, leading to a drastic reduction in regenerative potential and thereby driving the ageing process. Conversely, ageing also adversely alters the stem cell niche, disrupting the molecular pathways underlying stem cell homing, self-renewal, differentiation, and growth factor secretion, all of which are critical for tissue repair and regeneration. A holistic understanding of these molecular mechanisms, through empirical research and clinical trials, is essential for designing targeted therapies to modulate ageing and improve health parameters in older individuals.
Collapse
Affiliation(s)
- Jitendra Kumar Chaudhary
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India; Department of Zoology, Shivaji College, University of Delhi, New Delhi 110027, India.
| | - Ajay Kumar Danga
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India; National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Anita Kumari
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Akshay Bhardwaj
- Global Research Alliances, Ashoka University, Rajiv Gandhi Education City, Sonepat, Haryana 131029, India.
| | - Pramod C Rath
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
4
|
Kvach AY, Kutyumov VA, Starunov VV, Ostrovsky AN. Transcriptomic Landscape of Polypide Development in the Freshwater Bryozoan Cristatella mucedo: From Budding to Degeneration. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2025; 344:119-135. [PMID: 39831659 DOI: 10.1002/jez.b.23285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/26/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Colonial invertebrates consist of iterative semi-autonomous modules (usually termed zooids) whose lifespan is significantly shorter than that of the entire colony. Typically, module development begins with budding and ends with degeneration. Most studies on the developmental biology of colonial invertebrates have focused on blastogenesis, whereas the changes occurring throughout the entire zooidal life were examined only for a few tunicates. Here we provide the first description of transcriptomic changes during polypide development in the freshwater bryozoan Cristatella mucedo. For the first time for Bryozoa, we performed bulk RNA sequencing of six polypide stages in C. mucedo (buds, juvenile polypides, three mature stages, and degeneration stage) and generated a high-quality de novo reference transcriptome. Based on these data, we analyzed clusters of differentially expressed genes for enriched pathways and biological processes that may be involved in polypide budding, growth, active functioning, and degradation. Although stem cells have never been described in Bryozoa, our analysis revealed the expression of conservative "stemness" markers in developing buds and juvenile polypides. Our data also indicate that polypide degeneration is a complex regulated process involving autophagy and other types of programmed cell death. We hypothesize that the mTOR signaling pathway plays an important role in regulating the polypide lifespan.
Collapse
Affiliation(s)
- A Yu Kvach
- Department of Invertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Saint Petersburg, Russia
| | - V A Kutyumov
- Department of Invertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Saint Petersburg, Russia
| | - V V Starunov
- Department of Invertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Saint Petersburg, Russia
- Laboratory of Evolutionary Morphology, Zoological Institute, Russian Academy of Sciences, Saint Petersburg, Russia
| | - A N Ostrovsky
- Department of Invertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Saint Petersburg, Russia
- Department of Palaeontology, Faculty of Earth Sciences, Geography and Astronomy, University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Su R, Shen G, Xiao X, Zheng Y, Liu F, Chen D. Generation of a Novel Inducible and Dermal Papilla-Specific Wif1-CreER Knock-In Mouse Line for Hair Follicle Research. Exp Dermatol 2025; 34:e70109. [PMID: 40329691 DOI: 10.1111/exd.70109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 04/07/2025] [Accepted: 04/20/2025] [Indexed: 05/08/2025]
Abstract
Dermal papilla (DP) cells are essential niche cells that regulate hair follicle development, cycling and regeneration. Despite the establishment of several DP cell mouse lines in prior research, these tools are limited by incomplete specificity and spatiotemporal control. The Wnt inhibitory factor 1 (Wif1) has been identified as a DP signature gene. To address the need for precise labelling and manipulation of DP cells, we developed a novel genetic tool-Wif1-CreER knock-in mice. Using CRISPR/Cas9-mediated homologous recombination, the CreERT2 sequences were inserted into the endogenous Wif1 locus, under the control of the native promoter. PCR and sequencing analysis confirmed the accurate insertion of the CreERT2 sequence. Crossing Wif1-CreER mice with a reporter line demonstrated efficient and specific Cre recombinase activity in DP cells during anagen, catagen and telogen upon tamoxifen treatment across hair types. Importantly, DP-restricted labelling was confirmed by immunofluorescence and colocalised with Crabp1 and alkaline phosphatase (AP)-staining activity, exhibiting minimal to negligible expression in other tissues. This innovative mouse model overcomes the limitations of current tools and provides a valuable resource for advancing our understanding of hair biology and developing targeted therapies for hair-related disorders, offering unprecedented precision in the manipulation of dermal papilla cells.
Collapse
Affiliation(s)
- Rina Su
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Guangqian Shen
- Department of Breast Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xin Xiao
- National Institute of Biological Sciences, Beijing, China
| | - Yinghui Zheng
- National Institute of Biological Sciences, Beijing, China
| | - Fang Liu
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Daoming Chen
- National Institute of Biological Sciences, Beijing, China
| |
Collapse
|
6
|
Till NA, Ramanathan M, Bertozzi CR. Induced proximity at the cell surface. Nat Biotechnol 2025; 43:702-711. [PMID: 40140559 DOI: 10.1038/s41587-025-02592-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/17/2025] [Indexed: 03/28/2025]
Abstract
Molecular proximity is a governing principle of biology that is essential to normal and disease-related biochemical pathways. At the cell surface, protein-protein proximity regulates receptor activation, inhibition and protein recycling and degradation. Induced proximity is a molecular engineering principle in which bifunctional molecules are designed to bring two protein targets into close contact, inducing a desired biological outcome. Researchers use this engineering principle for therapeutic purposes and to interrogate fundamental biological mechanisms. This Review focuses on the use of induced proximity at the cell surface for diverse applications, such as targeted protein degradation, receptor inhibition and activating intracellular signaling cascades. We see a rich future for proximity-based modulation of cell surface protein activity both in basic and translational science.
Collapse
Affiliation(s)
- Nicholas A Till
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Muthukumar Ramanathan
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
7
|
Ahmad S, Hashim PK, Imajo M, Cheruthu NM, Takahashi K, Tanaka S, Nakamura T, Tamaoki N. Photoswitchable agonists for visible-light activation of the Wnt signaling pathway. Org Biomol Chem 2025; 23:4240-4245. [PMID: 40197693 DOI: 10.1039/d4ob01827c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Based on the known Wnt agonist BML-284, we designed and synthesized photoswitchable azo derivative compounds that can act as agonists for the Wnt signaling pathway. These photoswitchable agonists were shown to undergo reversible trans-cis isomerization upon being irradiated with visible light, but only the cis isomer was observed to activate the Wnt signaling pathway, using a luminescense-based reporter assay in cultured cells. One of the compounds, denoted as compound 2, showed ∼88% agonist activity after being subjected to visible light irradiation in comparison to the non-photoswitchable BML-284. We also were able to selectively activate the Wnt signaling pathway using 2 and light irradiation at a specific region of interest in a model cell culture system, highlighting the ability to achieve spatiotemporal regulation.
Collapse
Affiliation(s)
- Shifa Ahmad
- Research Institute for Electronic Science, Hokkaido University, Kita20, Nishi 10, Kita-ku, Sapporo, Hokkaido, 001-0020, Japan.
- Graduate School of Life Science, Hokkaido University, Kita 10, Nishi 8, Kita-ku, Sapporo, Hokkaido, 060-0810, Japan
| | - P K Hashim
- Research Institute for Electronic Science, Hokkaido University, Kita20, Nishi 10, Kita-ku, Sapporo, Hokkaido, 001-0020, Japan.
- Graduate School of Life Science, Hokkaido University, Kita 10, Nishi 8, Kita-ku, Sapporo, Hokkaido, 060-0810, Japan
| | - Masamichi Imajo
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21, W10, Kita-ku, Sapporo 001-0021, Japan
| | - Nusaiba Madappuram Cheruthu
- Research Institute for Electronic Science, Hokkaido University, Kita20, Nishi 10, Kita-ku, Sapporo, Hokkaido, 001-0020, Japan.
- Graduate School of Life Science, Hokkaido University, Kita 10, Nishi 8, Kita-ku, Sapporo, Hokkaido, 060-0810, Japan
| | - Kiyonori Takahashi
- Research Institute for Electronic Science, Hokkaido University, Kita20, Nishi 10, Kita-ku, Sapporo, Hokkaido, 001-0020, Japan.
- Graduate School of Environmental Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Shinya Tanaka
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21, W10, Kita-ku, Sapporo 001-0021, Japan
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15, W7, Kita-ku, Sapporo 060-8638, Japan
| | - Takayoshi Nakamura
- Research Institute for Electronic Science, Hokkaido University, Kita20, Nishi 10, Kita-ku, Sapporo, Hokkaido, 001-0020, Japan.
- Graduate School of Environmental Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Nobuyuki Tamaoki
- Research Institute for Electronic Science, Hokkaido University, Kita20, Nishi 10, Kita-ku, Sapporo, Hokkaido, 001-0020, Japan.
- Graduate School of Life Science, Hokkaido University, Kita 10, Nishi 8, Kita-ku, Sapporo, Hokkaido, 060-0810, Japan
| |
Collapse
|
8
|
Pang X, Pan Y, Wang M, Qiu S, He Y, Ren Y, Yu T, Yu S, Cui Y. Comparison of reproductive performance and functional analysis of spermatogenesis factors between domestic yak and semi-wild blood yak. BMC Genomics 2025; 26:418. [PMID: 40301732 PMCID: PMC12038992 DOI: 10.1186/s12864-025-11594-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 04/10/2025] [Indexed: 05/01/2025] Open
Abstract
This study investigates differences in reproductive performance, testicular histology, and transcriptomic profiles between male Subei (SB; semi-wild) yaks and two domestic yaks, Gannan (GN) and Qinghai (QH). Key metrics including mating age, utilization time, breeding capacity, morphometric traits, and testicular indices were analyzed. SB yaks exhibited superior reproductive metrics, including earlier sexual maturity, prolonged utilization periods, and enhanced breeding capacity compared to GN and QH (P < 0.05). Morphologically, SB yaks demonstrated significantly greater body weight, and testicular dimensions. Compared with GN and QH yaks, the seminiferous tubules of SB yaks exhibited significantly larger spermatogenic cells and luminal cavities, along with a notably higher sperm density within the luminal cavity. Transcriptomic analysis identified 2,403 and 4,428 differentially expressed genes (DEGs) in GN vs. SB and QH vs. SB comparisons, respectively. Eight key genes (TPPP3, SMAD3, PAFAH1B3, BMP7, ARSA, CTNNB1, SMAD4, STAT3) and three pathways (Hippo, pluripotency regulation, TGF-β) were implicated in testicular development and spermatogenesis. These findings underscore the genetic and physiological advantages of SB yaks, offering insights for enhancing male yak reproductive performance.
Collapse
Affiliation(s)
- Xin Pang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Gansu Innovation Centre for Livestock Embryo Engineering and Technology, Lanzhou, China
| | - Yangyang Pan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Gansu Innovation Centre for Livestock Embryo Engineering and Technology, Lanzhou, China
| | - Meng Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Gansu Innovation Centre for Livestock Embryo Engineering and Technology, Lanzhou, China
| | - Shantong Qiu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Gansu Innovation Centre for Livestock Embryo Engineering and Technology, Lanzhou, China
| | - Yulong He
- Jiuquan Animal Husbandry and Veterinary Medicine General Station, Jiuquan, China
| | - Yuchun Ren
- Central Agricultural Radio and Television School Tianzhu County Branch, Wuwei, China
| | - Tianjun Yu
- Subei Mongolian Autonomous County Animal Husbandry and Veterinary Technical Service Center, Jiuquan, China
| | - Sijiu Yu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China.
- Gansu Innovation Centre for Livestock Embryo Engineering and Technology, Lanzhou, China.
| | - Yan Cui
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China.
- Gansu Innovation Centre for Livestock Embryo Engineering and Technology, Lanzhou, China.
| |
Collapse
|
9
|
Gao H, Wang L, Lyu Y, Jin H, Lin Z, Kang Y, Li Z, Zhang X, Jiang Y, Zhang G, Tao Z, Zhang X, Yang B, Bai X, Ma X, Liu S, Jiang J. The P2X7R/NLRP3 inflammasome axis suppresses enthesis regeneration through inflammatory and metabolic macrophage-stem cell cross-talk. SCIENCE ADVANCES 2025; 11:eadr4894. [PMID: 40279432 PMCID: PMC12024643 DOI: 10.1126/sciadv.adr4894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 03/21/2025] [Indexed: 04/27/2025]
Abstract
The regeneration of the enthesis remains a formidable challenge in regenerative medicine. However, key regulators underlying unsatisfactory regeneration remain poorly understood. This study reveals that the purinergic receptor P2X7 (P2X7R)/Nod-like receptor family protein 3 (NLRP3) inflammasome axis suppresses enthesis regeneration by amplifying IL-1β-mediated inflammatory cross-talk and suppressing docosatrienoic acid (DTA) metabolic cross-talk. NLRP3 inflammasomes were activated in macrophages following enthesis injury, thereby impairing the histological and functional recovery of the injured enthesis. Single-cell RNA sequencing (scRNA-seq) indicated that Nlrp3 knockout attenuated pathological inflammation and ameliorated the detrimental effects of IL-1β signaling cross-talk. Furthermore, NLRP3 inflammasomes suppressed the secretion of anti-inflammatory cytokines (IL-10 and IL-13) and DTA. The NLRP3 inflammasome-mediated secretome reduced differentiation and migration of stem cells. Neutralizing IL-1β or replenishing docosatrienoic acid accelerated enthesis regeneration. Moreover, conditional knockout of P2rx7 in myeloid cells attenuated NLRP3 inflammasome activation and facilitated enthesis regeneration. This study demonstrates that the P2X7R/NLRP3 inflammasome axis represents a promising therapeutic target for enthesis repair.
Collapse
Affiliation(s)
- Haihan Gao
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, Shanghai 200020, China
| | - Liren Wang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, Shanghai 200020, China
| | - Yangbao Lyu
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Haocheng Jin
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zhiqi Lin
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yuhao Kang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ziyun Li
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xueying Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yuhan Jiang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Guoyang Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zaijin Tao
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xiaofeng Zhang
- Department of Orthopedic Surgery, Jinshan Branch of Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201500, China
| | - Bin Yang
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xingyu Bai
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xin Ma
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Shen Liu
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jia Jiang
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| |
Collapse
|
10
|
Sharma A, Zalejski J, Bendre SV, Kavrokova S, Hasdemir HS, Ozgulbas DG, Sun J, Pathmasiri KC, Shi R, Aloulou A, Berkley K, Delisle CF, Wang Y, Weisser E, Buweneka P, Pierre-Jacques D, Mukherjee S, Abbasi DA, Lee D, Wang B, Gevorgyan V, Cologna SM, Tajkhorshid E, Nelson ER, Cho W. Cholesterol-targeting Wnt-β-catenin signaling inhibitors for colorectal cancer. Nat Chem Biol 2025:10.1038/s41589-025-01870-y. [PMID: 40240631 DOI: 10.1038/s41589-025-01870-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/28/2025] [Indexed: 04/18/2025]
Abstract
Most persons with colorectal cancer (CRC) carry adenomatous polyposis coli (APC) truncation leading to aberrant Wnt-β-catenin signaling; however, effective targeted therapy for them is lacking as the mechanism by which APC truncation drives CRC remains elusive. Here, we report that the cholesterol level in the inner leaflet of the plasma membrane (IPM) is elevated in all tested APC-truncated CRC cells, driving Wnt-independent formation of Wnt signalosomes through Dishevelled (Dvl)-cholesterol interaction. Cholesterol-Dvl interaction inhibitors potently blocked β-catenin signaling in APC-truncated CRC cells and suppressed their viability. Because of low IPM cholesterol level and low Dvl expression and dependence, normal cells including primary colon epithelial cells were not sensitive to these inhibitors. In vivo testing with a xenograft mouse model showed that our inhibitors effectively suppressed truncated APC-driven tumors without causing intestinal toxicity. Collectively, these results suggest that the most common type of CRC could be effectively and safely treated by blocking the cholesterol-Dvl-β-catenin signaling axis.
Collapse
Affiliation(s)
- Ashutosh Sharma
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Julian Zalejski
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Shruti Vijay Bendre
- Department of Molecular and Integrative Physiology, Cancer Center at Illinois, Beckman Institute for Advanced Science and Technology, Carl R. Woese Institute for Genomic Biology- Anticancer Discovery from Pets to People, Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Simona Kavrokova
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Hale Siir Hasdemir
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Defne Gorgun Ozgulbas
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Jiachen Sun
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | | | - Ruicheng Shi
- Department of Comparative Biosciences, Division of Nutritional Sciences, Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Ahmed Aloulou
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Kyli Berkley
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Charles F Delisle
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Young Wang
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Erin Weisser
- Department of Molecular and Integrative Physiology, Cancer Center at Illinois, Beckman Institute for Advanced Science and Technology, Carl R. Woese Institute for Genomic Biology- Anticancer Discovery from Pets to People, Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Pawanthi Buweneka
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | | | - Sayandeb Mukherjee
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Diana A Abbasi
- Department of Neurogenetics and Translational Neuroscience, Rush University, Chicago, IL, USA
| | - Daesung Lee
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | - Bo Wang
- Department of Comparative Biosciences, Division of Nutritional Sciences, Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | | | | | - Emad Tajkhorshid
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, Cancer Center at Illinois, Beckman Institute for Advanced Science and Technology, Carl R. Woese Institute for Genomic Biology- Anticancer Discovery from Pets to People, Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Wonhwa Cho
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
11
|
Zhang S, Cao Y, Huang Y, Zhang X, Mou C, Qin T, Chen Z, Bao W. Abortive PDCoV infection triggers Wnt/β-catenin pathway activation, enhancing intestinal stem cell self-renewal and promoting chicken resistance. J Virol 2025; 99:e0013725. [PMID: 40135895 PMCID: PMC11998530 DOI: 10.1128/jvi.00137-25] [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: 01/24/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging coronavirus causing economic losses to swine industries worldwide. PDCoV can infect chickens under laboratory conditions, usually with no symptoms or mild symptoms, and may cause outbreaks in backyard poultry and wildfowl, posing a potential risk of significant economic loss to the commercial poultry industry. However, the reasons for such a subdued reaction after infection are not known. Here, using chicken intestinal organoid monolayers, we found that although PDCoV infects them nearly as well as porcine intestinal organoid monolayers, infection did not result in detectable amounts of progeny virus. In ex vivo and in vivo experiments using chickens, PDCoV infection failed to initiate interferon and inflammatory responses. Additionally, infection did not result in a disrupted intestinal barrier nor a reduced number of goblet cells and mucus secretion, as in pigs. In fact, the number of goblet cells increased as did the secreted mucus, thereby providing an enhanced protective barrier. Ex vivo PDCoV infection in chicken triggered activation of the Wnt/β-catenin pathway with the upregulation of Wnt/β-catenin pathway genes (Wnt3a, Lrp5, β-catenin, and TCF4) and Wnt target genes (Lgr5, cyclin D1, and C-myc). This activation stimulates the self-renewal of intestinal stem cells (ISCs), accelerating ISC-mediated epithelial regeneration by significant up-regulation of PCNA (transiently amplifying cells), BMI1 (ISCs), and Lyz (Paneth cells). Our data demonstrate that abortive infection of PDCoV in chicken cells activates the Wnt/β-catenin pathway, which facilitates the self-renewal and proliferation of ISCs, contributing to chickens' resistance to PDCoV infection.IMPORTANCEThe intestinal epithelium is the main target of PDCoV infection and serves as a physical barrier against pathogens. Additionally, ISCs are charged with tissue repair after injury, and promoting rapid self-renewal of intestinal epithelium will help to re-establish the physical barrier and maintain intestinal health. We found that PDCoV infection in chicken intestinal organoid monolayers resulted in abortive infection and failed to produce infectious virions, disrupt the intestinal barrier, reduce the number of goblet cells and mucus secretion, and induce innate immunity, but rather increased goblet cell numbers and mucus secretion. Abortive PDCoV infection activated the Wnt/β-catenin pathway, enhancing ISC renewal and accelerating the renewal and replenishment of shed PDCoV-infected intestinal epithelial cells, thereby enhancing chicken resistance to PDCoV infection. This study provides novel insights into the mechanisms underlying the mild or asymptomatic response to PDCoV infection in chickens, which is critical for understanding the virus's potential risks to the poultry industry.
Collapse
Affiliation(s)
- Shuai Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanan Cao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanjie Huang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Xueli Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Chunxiao Mou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Tao Qin
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhenhai Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
12
|
Wu T, Zhang X, Zeng X, Liu Y, Wang L, Huang Y. Wnt7a can upregulate cell adhesion and migration related genes expression and facilitate the repair of corneal epithelial cells after injury. Int Ophthalmol 2025; 45:149. [PMID: 40192882 DOI: 10.1007/s10792-025-03506-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 03/09/2025] [Indexed: 05/17/2025]
Abstract
PURPOSE To investigate the role of Wnt7a in corneal epithelial repair and its underlying mechanisms. METHODS Immunohistochemistry and immunofluorescence were used to assess Wnt7a expression in mouse corneas under normal and injury conditions. Human corneal epithelial cells (HCECs) were treated with Wnt7a siRNA or recombinant human Wnt7a (rhWnt7a) to evaluate proliferation (CCK-8 assay) and migration (scratch assay). Transcriptome sequencing and western blotting were performed to identify Wnt7a-regulated pathways and proteins. RESULTS Under normal conditions, Wnt7a was predominantly localized to the corneal limbus basal cells. Following corneal injury, its expression significantly increased in central corneal epithelial cells and co-localized with nuclei during repair. Wnt7a siRNA suppressed HCEC proliferation and migration, while rhWnt7a enhanced proliferation. Transcriptome analysis revealed upregulation of cell adhesion-related genes (e.g., FN1, ITGBs, LAMs), particularly fibronectin (FN), validated by increased FN protein levels after rhWnt7a treatment. Pathway enrichment implicated PI3K/Akt, Wnt signaling, and ECM-receptor interactions. CONCLUSION Wnt7a promotes corneal epithelial repair by enhancing migration and proliferation, primarily through upregulating fibronectin and ECM-related pathways. These findings highlight Wnt7a as a potential therapeutic target for accelerating corneal wound healing.
Collapse
Affiliation(s)
- Tengyun Wu
- General Hospital of Chinese PLA, Beijing, 100038, China
- Air Force Medical Center of Chinese PLA, Beijing, 100142, China
| | - Xianxie Zhang
- Department of Pharmaceutical Science, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xiangwen Zeng
- General Hospital of Chinese PLA, Beijing, 100038, China
| | - Yong Liu
- Air Force Medical Center of Chinese PLA, Beijing, 100142, China
| | - Liqiang Wang
- General Hospital of Chinese PLA, Beijing, 100038, China.
| | - Yifei Huang
- General Hospital of Chinese PLA, Beijing, 100038, China.
| |
Collapse
|
13
|
Chen Y, Hou Y, Chen J, Bai J, Du L, Qiu C, Qi H, Liu X, Huang J. Construction of Large-Scale Bioengineered Hair Germs and In Vivo Transplantation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416361. [PMID: 40042061 PMCID: PMC12021125 DOI: 10.1002/advs.202416361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/21/2025] [Indexed: 04/26/2025]
Abstract
Hair follicle (HF) regeneration technology holds promise for treating hair loss, but creating a biomimetic structure that mimics the natural follicle microenvironment remains challenging. Here a novel bioengineered hair germ (BHG) is developed using thermodynamically incompatible mucopolysaccharides to enhance HF regeneration efficiency. Mucopolysaccharide-based hydrogels are synthesized by grafting amino and diethylamino groups (dihydroxyphenylalanine-grafted hyaluronic acid (HME) hydrogels) for rapid gelation and strong wetting adhesion. Dual-layered microspheres are fabricated using a co-flow microfluidic system, with HME as the outer shell and gelatin methacrylate (GelMA) as the core, achieving thermodynamic incompatibility. The Wnt3a protein is encapsulated for sustained release. RNA sequencing, reverse transcription quantitative polymerase chain reaction (RT-qPCR), and functional validation are used to study the molecular mechanisms of HF regeneration. Results show that HME hydrogels exhibit excellent adhesion, shear-thinning behavior, and biocompatibility. The microspheres release Wnt3a for up to 9 days, with high-throughput sequencing revealing upregulation of HF regeneration genes like Ctnnb1 and Lef1, and activation of the Wnt signaling pathway, while hypoxia-related genes such as Hif-1ɑ are downregulated. Pathway enrichment analyses confirm the enrichment of HF regeneration pathways. In conclusion, the HME-based BHG microspheres effectively promote in vivo HF regeneration, offering a promising solution for hair loss treatment and regeneration.
Collapse
Affiliation(s)
- Yangpeng Chen
- Department of Plastic and Aesthetic SurgeryDepartment of HematologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yuhui Hou
- Department of Plastic and Aesthetic SurgeryDepartment of HematologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Jiejian Chen
- Department of Plastic and Aesthetic SurgeryDepartment of HematologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- Department of Medical OncologyGuangzhou First People's HospitalGuangzhou Medical UniversityGuangdong510180China
| | - Jiaojiao Bai
- Department of Plastic and Aesthetic SurgeryDepartment of HematologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- Department of HaematologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)GuangzhouGuangdong510030China
| | - Lijuan Du
- Department of Plastic and Aesthetic SurgeryDepartment of HematologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Chen Qiu
- Department of OncologyShanghai General Hospital650 Xinsongjiang RoadSongjiang DistrictShanghai201620China
| | - Hanzhou Qi
- Department of Plastic and Aesthetic SurgeryDepartment of HematologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Xuanbei Liu
- Department of Plastic and Aesthetic SurgeryDepartment of HematologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Junfei Huang
- Department of Plastic and Aesthetic SurgeryDepartment of HematologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| |
Collapse
|
14
|
Wong GP, Hartmann S, Nonn O, Cannon P, Nguyen TV, Kandel M, de Alwis N, Murphy CN, Pritchard N, Dechend R, Hannan NJ, Tong S, Simmons DG, Kaitu'u-Lino TJ. Stem Cell Markers LGR5, LGR4 and Their Immediate Signalling Partners are Dysregulated in Preeclampsia. Stem Cell Rev Rep 2025; 21:872-896. [PMID: 39688759 DOI: 10.1007/s12015-024-10831-2] [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] [Accepted: 11/27/2024] [Indexed: 12/18/2024]
Abstract
Leucine-rich repeat-containing G protein-coupled receptors 5/4 (LGR5/LGR4) are critical stem cell markers in epithelial tissues including intestine. They agonise wingless-related integration site (WNT) signalling. Until now, LGR5/LGR4 were uncharacterised in placenta, where analogous functions may exist. We characterised LGR5/LGR4, their ligands/targets in human placenta, with further assessments on dysregulation in preeclampsia/fetal growth restriction (FGR). LGR5 mRNA was unaltered in first trimester (n = 11), preterm (n = 9) and term (n = 11) placental lysate. LGR5 was enriched in human trophoblast stem cells (hTSCs) and downregulated with differentiation to extravillous trophoblasts (p < 0.0215) and syncytiotrophoblasts (p < 0.0350). In situ hybridisation localised LGR5 to unique, proliferative MKI67 + mononuclear trophoblasts underlying syncytium which concurred with proposed progenitor identities in single-cell transcriptomics. LGR5 expression was significantly reduced in placentas from early-onset preeclampsia (p < 0.0001, n = 81 versus n = 19 controls), late-onset preeclampsia (p = 0.0046, n = 20 versus n = 33 controls) and FGR (p = 0.0031, n = 34 versus n = 17 controls). LGR4 was elevated in first trimester versus preterm and term placentas (p = 0.0412), in placentas with early-onset preeclampsia (p = 0.0148) and in FGR (p = 0.0417). Transcriptomic analysis and in vitro hTSC differentiation to both trophoblast lineages suggested LGR4 increases with differentiation. Single-nucleus RNA sequencing of placental villous samples supported LGR5 and LGR4 localisation findings. Hypoxia/proinflammatory cytokine treatment modelling elements experienced by the placenta in placental insufficiency pathogenesis did not significantly alter LGR5/LGR4. Ligands R-spondins 1/3/4, and neutralising targets ring finger protein 43 (RNF43) and zinc and ring finger 3 (ZNRF3) were also reduced in placentas from preeclamptic pregnancies. This study is the first to describe LGR5/LGR4 and their signalling partner expression in human placenta. Their dysregulations in the preeclamptic placenta allude to disruptions to integral trophoblast stem cell function/differentiation that may occur during placental development related to WNT signalling.
Collapse
Affiliation(s)
- Georgia P Wong
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia.
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia.
| | - Sunhild Hartmann
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
- Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and Charitè Campus Buch, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site, Berlin, Germany
| | - Olivia Nonn
- Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and Charitè Campus Buch, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site, Berlin, Germany
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Ping Cannon
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Tuong-Vi Nguyen
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Manju Kandel
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Natasha de Alwis
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Ciara N Murphy
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Natasha Pritchard
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Ralf Dechend
- Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and Charitè Campus Buch, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site, Berlin, Germany
- Department of Cardiology and Nephrology, HELIOS Klinikum, Berlin Buch, Germany
| | - Natalie J Hannan
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Stephen Tong
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - David G Simmons
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
15
|
Fang F, Li G, Li X, Wu J, Liu Y, Xin H, Wang Z, Fang J, Jiang Y, Qian W, Hou X, Song J. Piezo1 regulates colon stem cells to maintain epithelial homeostasis through SCD1-Wnt-β-catenin and programming fatty acid metabolism. Cell Rep 2025; 44:115400. [PMID: 40080500 DOI: 10.1016/j.celrep.2025.115400] [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: 09/24/2024] [Revised: 01/05/2025] [Accepted: 02/14/2025] [Indexed: 03/15/2025] Open
Abstract
Piezo1, which maintains the integrity and function of the intestinal epithelial barrier, is essential for colonic epithelial homeostasis. However, whether and how Piezo1 regulates colon stem cell fate remains unclear. Here, we show that Piezo1 inhibition promotes colon stem cell proliferation. Mechanistically, stearoyl-CoA 9-desaturase 1 (SCD1) is downstream of Piezo1 to affect colon stem cell stemness by acting on the Wnt-β-catenin pathway. For mice, the altered colon stem cell stemness after Piezo1 knockdown and activation was accompanied by a reprogrammed fatty acid (FA) metabolism in colon crypts. Notably, we found that GsMTX4 protects injured colon stem cell stemness in mouse and human colitis organoids. Our results elucidated the role of Piezo1 in regulating normal and postinjury colon stem cell fates through SCD1-Wnt-β-catenin and the SCD1-mediated FA desaturation process. These results provide fresh perspectives on the mechanical factors regulating colon stem cell fate and therapeutic strategies for related intestinal diseases.
Collapse
Affiliation(s)
- Feifei Fang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gangping Li
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xueyan Li
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiandi Wu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ying Liu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Haoren Xin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhe Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jianhua Fang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yudong Jiang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wei Qian
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaohua Hou
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jun Song
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
16
|
Zheng Z, Su Z, Zhang W. Melatonin's Role in Hair Follicle Growth and Development: A Cashmere Goat Perspective. Int J Mol Sci 2025; 26:2844. [PMID: 40243438 PMCID: PMC11988770 DOI: 10.3390/ijms26072844] [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: 02/03/2025] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Hair follicles, unique skin appendages, undergo cyclic phases (anagen, catagen, telogen) governed by melatonin and associated molecular pathways. Melatonin, synthesized in the pineal gland, skin, and gut, orchestrates these cycles through antioxidant activity and signaling cascades (e.g., Wnt, BMP). This review examines melatonin's biosynthesis across tissues, its regulation of cashmere growth patterns, and its interplay with non-coding RNAs and the gut-skin axis. Recent advances highlight melatonin's dual role in enhancing antioxidant capacity (via Keap1-Nrf2) and modulating gene expression (e.g., Wnt10b, CTNNB1) to promote hair follicle proliferation. By integrating multi-omics insights, we construct a molecular network of melatonin's regulatory mechanisms, offering strategies to improve cashmere yield and quality while advancing therapies for human alopecia.
Collapse
Affiliation(s)
| | | | - Wei Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Z.Z.); (Z.S.)
| |
Collapse
|
17
|
Barber AG, Quintero CM, Hamilton M, Rajbhandari N, Sasik R, Zhang Y, Kim C, Husain H, Sun X, Reya T. Regulation of lung cancer initiation and progression by the stem cell determinant Musashi. eLife 2025; 13:RP97021. [PMID: 40047406 PMCID: PMC11884785 DOI: 10.7554/elife.97021] [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] [Indexed: 03/09/2025] Open
Abstract
Despite advances in therapeutic approaches, lung cancer remains the leading cause of cancer-related deaths. To understand the molecular programs underlying lung cancer initiation and maintenance, we focused on stem cell programs that are normally extinguished with differentiation but can be reactivated during oncogenesis. Here, we have used extensive genetic modeling and patient-derived xenografts (PDXs) to identify a dual role for Msi2: as a signal that acts initially to sensitize cells to transformation, and subsequently to drive tumor propagation. Using Msi reporter mice, we found that Msi2-expressing cells were marked by a pro-oncogenic landscape and a preferential ability to respond to Ras and p53 mutations. Consistent with this, genetic deletion of Msi2 in an autochthonous Ras/p53-driven lung cancer model resulted in a marked reduction of tumor burden, delayed progression, and a doubling of median survival. Additionally, this dependency was conserved in human disease as inhibition of Msi2 impaired tumor growth in PDXs. Mechanistically, Msi2 triggered a broad range of pathways critical for tumor growth, including several novel effectors of lung adenocarcinoma. Collectively, these findings reveal a critical role for Msi2 in aggressive lung adenocarcinoma, lend new insight into the biology of this disease, and identify potential new therapeutic targets.
Collapse
Affiliation(s)
- Alison G Barber
- Department of Pharmacology and Medicine, University of California San Diego School of MedicineLa JollaUnited States
- Moores Cancer Center, University of California San Diego School of MedicineLa JollaUnited States
| | - Cynthia M Quintero
- Department of Pharmacology and Medicine, University of California San Diego School of MedicineLa JollaUnited States
- Moores Cancer Center, University of California San Diego School of MedicineLa JollaUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical CenterNew YorkUnited States
- Department of Physiology and Cellular Biophysics, Columbia University Medical CenterNew YorkUnited States
| | - Michael Hamilton
- Department of Pharmacology and Medicine, University of California San Diego School of MedicineLa JollaUnited States
- Moores Cancer Center, University of California San Diego School of MedicineLa JollaUnited States
| | - Nirakar Rajbhandari
- Department of Pharmacology and Medicine, University of California San Diego School of MedicineLa JollaUnited States
- Moores Cancer Center, University of California San Diego School of MedicineLa JollaUnited States
| | - Roman Sasik
- Center for Computational Biology and Bioinformatics, University of California San Diego School of MedicineLa JollaUnited States
| | - Yan Zhang
- Department of Pediatrics, University of California, San DiegoLa JollaUnited States
| | - Carla Kim
- Stem Cell Program, Division of Hematology/Oncology and Division of Respiratory Disease, Boston Children’s HospitalBostonUnited States
- Department of Genetics, Harvard Medical SchoolBostonUnited States
- Harvard Stem Cell InstituteCambridgeUnited States
| | - Hatim Husain
- Moores Cancer Center, University of California San Diego School of MedicineLa JollaUnited States
| | - Xin Sun
- Department of Pediatrics, University of California, San DiegoLa JollaUnited States
| | - Tannishtha Reya
- Department of Pharmacology and Medicine, University of California San Diego School of MedicineLa JollaUnited States
- Moores Cancer Center, University of California San Diego School of MedicineLa JollaUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical CenterNew YorkUnited States
- Department of Physiology and Cellular Biophysics, Columbia University Medical CenterNew YorkUnited States
| |
Collapse
|
18
|
Rhim WK, Woo J, Kim JY, Lee EH, Cha SG, Kim DS, Baek SW, Park CG, Kim BS, Kwon TG, Han DK. Multiplexed PLGA scaffolds with nitric oxide-releasing zinc oxide and melatonin-modulated extracellular vesicles for severe chronic kidney disease. J Adv Res 2025; 69:75-89. [PMID: 38537702 PMCID: PMC11954823 DOI: 10.1016/j.jare.2024.03.018] [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: 11/13/2023] [Revised: 03/15/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION With prevalence of chronic kidney disease (CKD) in worldwide, the strategies to recover renal function via tissue regeneration could provide alternatives to kidney replacement therapies. However, due to relatively low reproducibility of renal basal cells and limited bioactivities of implanted biomaterials along with the high probability of substance-inducible inflammation and immunogenicity, kidney tissue regeneration could be challenging. OBJECTIVES To exclude various side effects from cell transplantations, in this study, we have induced extracellular vesicles (EVs) incorporated cell-free hybrid PMEZ scaffolds. METHODS Hybrid PMEZ scaffolds incorporating essential bioactive components, such as ricinoleic acid grafted Mg(OH)2 (M), extracellular matrix (E), and alpha lipoic acid-conjugated ZnO (Z) based on biodegradable porous PLGA (P) platform was successfully manufactured. Consecutively, for functional improvements, melatonin-modulated extracellular vesicles (mEVs), derived from the human umbilical cord MSCs in chemically defined media without serum impurities, were also loaded onto PMEZ scaffolds to construct the multiplexed PMEZ/mEV scaffold. RESULTS With functionalities of Mg(OH)2 and extracellular matrix-loaded PLGA scaffolds, the continuous nitric oxide-releasing property of modified ZnO and remarkably upregulated regenerative functionalities of mEVs showed significantly enhanced kidney regenerative activities. Based on these, the structural and functional restoration has been practically achieved in 5/6 nephrectomy mouse models that mimicked severe human CKD. CONCLUSION Our study has proved the combinatory bioactivities of the biodegradable PLGA-based multiplexed scaffold for kidney tissue regeneration in 5/6 nephrectomy mouse representing a severe CKD model. The optimal microenvironments for the morphogenetic formations of renal tissues and functional restorations have successfully achieved the combinatory bioactivities of remarkable components for PMEZ/mEV, which could be a promising therapeutic alternative for CKD treatment.
Collapse
Affiliation(s)
- Won-Kyu Rhim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Jiwon Woo
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Jun Yong Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea; Department of Biomedical Engineering and Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea; Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Eun Hye Lee
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea
| | - Seung-Gyu Cha
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea; Department of Biomedical Engineering and Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea; Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering and Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea; Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Bum Soo Kim
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea; Department of Urology, School of Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea
| | - Tae Gyun Kwon
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea; Department of Urology, School of Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea.
| |
Collapse
|
19
|
An P, Tong Y, Mu R, Han L. Wnt-Regulated Therapeutics for Blood-Brain Barrier Modulation and Cancer Therapy. Bioconjug Chem 2025; 36:136-145. [PMID: 39680846 DOI: 10.1021/acs.bioconjchem.4c00537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
The Wnt signaling pathway has a significant regulatory part in tissue development and homeostasis. Dysregulation of the Wnt signaling pathway has been associated with many diseases including cancers and various brain diseases, making this signaling pathway a promising therapeutic target for these diseases. In this review, we describe the roles of the Wnt signaling pathway in the blood-brain barrier (BBB) in intracranial tumors and peripheral tumors, from preclinical and clinical perspectives, introduce Wnt-regulated therapeutics including various types of drugs and nanomedicines as BBB modulators for brain-oriented drug delivery and as therapeutic drugs for cancer treatments, and finally discuss limitations and future perspectives for Wnt-regulated therapeutics.
Collapse
Affiliation(s)
- Pei An
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yang Tong
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Rui Mu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Liang Han
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China
| |
Collapse
|
20
|
Verona F, Di Bella S, Schirano R, Manfredi C, Angeloro F, Bozzari G, Todaro M, Giannini G, Stassi G, Veschi V. Cancer stem cells and tumor-associated macrophages as mates in tumor progression: mechanisms of crosstalk and advanced bioinformatic tools to dissect their phenotypes and interaction. Front Immunol 2025; 16:1529847. [PMID: 39981232 PMCID: PMC11839637 DOI: 10.3389/fimmu.2025.1529847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/17/2025] [Indexed: 02/22/2025] Open
Abstract
Cancer stem cells (CSCs) are a small subset within the tumor mass significantly contributing to cancer progression through dysregulation of various oncogenic pathways, driving tumor growth, chemoresistance and metastasis formation. The aggressive behavior of CSCs is guided by several intracellular signaling pathways such as WNT, NF-kappa-B, NOTCH, Hedgehog, JAK-STAT, PI3K/AKT1/MTOR, TGF/SMAD, PPAR and MAPK kinases, as well as extracellular vesicles such as exosomes, and extracellular signaling molecules such as cytokines, chemokines, pro-angiogenetic and growth factors, which finely regulate CSC phenotype. In this scenario, tumor microenvironment (TME) is a key player in the establishment of a permissive tumor niche, where CSCs engage in intricate communications with diverse immune cells. The "oncogenic" immune cells are mainly represented by B and T lymphocytes, NK cells, and dendritic cells. Among immune cells, macrophages exhibit a more plastic and adaptable phenotype due to their different subpopulations, which are characterized by both immunosuppressive and inflammatory phenotypes. Specifically, tumor-associated macrophages (TAMs) create an immunosuppressive milieu through the production of a plethora of paracrine factors (IL-6, IL-12, TNF-alpha, TGF-beta, CCL1, CCL18) promoting the acquisition by CSCs of a stem-like, invasive and metastatic phenotype. TAMs have demonstrated the ability to communicate with CSCs via direct ligand/receptor (such as CD90/CD11b, LSECtin/BTN3A3, EPHA4/Ephrin) interaction. On the other hand, CSCs exhibited their capacity to influence immune cells, creating a favorable microenvironment for cancer progression. Interestingly, the bidirectional influence of CSCs and TME leads to an epigenetic reprogramming which sustains malignant transformation. Nowadays, the integration of biological and computational data obtained by cutting-edge technologies (single-cell RNA sequencing, spatial transcriptomics, trajectory analysis) has significantly improved the comprehension of the biunivocal multicellular dialogue, providing a comprehensive view of the heterogeneity and dynamics of CSCs, and uncovering alternative mechanisms of immune evasion and therapeutic resistance. Moreover, the combination of biology and computational data will lead to the development of innovative target therapies dampening CSC-TME interaction. Here, we aim to elucidate the most recent insights on CSCs biology and their complex interactions with TME immune cells, specifically TAMs, tracing an exhaustive scenario from the primary tumor to metastasis formation.
Collapse
Affiliation(s)
- Francesco Verona
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, Italy
| | - Sebastiano Di Bella
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, Italy
| | - Roberto Schirano
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - Camilla Manfredi
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - Francesca Angeloro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Giulia Bozzari
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, Italy
| | - Matilde Todaro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
- Azienda Ospedaliera Universitaria Policlinico “Paolo Giaccone” (AOUP), Palermo, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
- Istituto Pasteur, Fondazione Cenci-Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Giorgio Stassi
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, Italy
| | - Veronica Veschi
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| |
Collapse
|
21
|
Kim J, Jeon YJ, Chang IY, Lee JH, You HJ. Disruption of the β-catenin destruction complex via Ephexin1-Axin1 interaction promotes colorectal cancer proliferation. Exp Mol Med 2025; 57:151-166. [PMID: 39741188 PMCID: PMC11799323 DOI: 10.1038/s12276-024-01381-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/16/2024] [Accepted: 10/20/2024] [Indexed: 01/02/2025] Open
Abstract
Wnt signaling is essential for cell growth and tumor formation and is abnormally activated in colorectal cancer (CRC), contributing to tumor progression; however, the specific role and regulatory mechanisms involved in tumor development remain unclear. Here, we show that Ephexin1, a guanine nucleotide exchange factor, is significantly overexpressed in CRC and is correlated with increased Wnt/β-catenin pathway activity. Through comprehensive analysis, including RNA sequencing data from TCGA and functional assays, we observed that Ephexin1 promotes tumor proliferation and migration by activating the Wnt/β-catenin pathway. This effect was mediated by the interaction of Ephexin1 with Axin1, a critical component of the β-catenin destruction complex, which in turn enhanced the stability and activity of β-catenin in signaling pathways critical for tumor development. Importantly, our findings also suggest that targeting Ephexin1 may increase the efficacy of Wnt/β-catenin pathway inhibitors in CRC treatment. These findings highlight the potential of targeting Ephexin1 as a strategy for developing effective treatments for CRC, suggesting a novel and promising approach to therapy aimed at inhibiting cancer progression.
Collapse
Affiliation(s)
- Jeeho Kim
- Laboratory of Genomic Instability and Cancer Therapeutics, Gwangju, South Korea
- Department of Pharmacology, Gwangju, South Korea
| | | | | | - Jung-Hee Lee
- Laboratory of Genomic Instability and Cancer Therapeutics, Gwangju, South Korea.
- Department of Cellular and Molecular Medicine, Chosun University School of Medicine, 375 Seosuk-dong, Gwangju, 501-759, South Korea.
| | - Ho Jin You
- Laboratory of Genomic Instability and Cancer Therapeutics, Gwangju, South Korea.
- Department of Pharmacology, Gwangju, South Korea.
| |
Collapse
|
22
|
Canosa S, Silvestris E, Carosso AR, Ruffa A, Evangelisti B, Gennarelli G, Cormio G, Loizzi V, Rolfo A, Benedetto C, Revelli A. Ovarian Stem Cells: Will the Dream of Neo-Folliculogenesis After Birth Become Real? Obstet Gynecol Surv 2025; 80:112-120. [PMID: 39924337 DOI: 10.1097/ogx.0000000000001360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Importance Ovarian stem cells (OSCs) represent a promising tool in reproductive medicine, particularly for the treatment of premature ovarian failure and fertility preservation. Objectives Herein, we summarize the main characteristics of adult stem cells, their status, needs, and new challenges in the application in reproductive medicine. Evidence Acquisition Clinical studies have shown that OSCs transplantation can restore ovarian function and stimulate neo-folliculogenesis in patients with premature ovarian failure, enabling them to conceive naturally or through in vitro fertilization techniques. Moreover, OSCs gained increasing interest as a chance to preserve fertility in cancer patients undergoing gonadotoxic treatments affecting their fertility, as chemotherapy or radiotherapy. Results The recruitment of OSCs from fresh or thawed ovarian fragments coupled with their capability to differentiate in vitro to mature oocytes could provide a novel opportunity to verify their suitability to be expanded in vitro as oocyte like cells. Conclusions and Relevance Research into OSCs and their applications in reproductive medicine is still in its infancy, but the results so far are promising and offer new possibilities for patients suffering from premature ovarian failure or cancer.
Collapse
Affiliation(s)
- Stefano Canosa
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant'Anna Hospital, University of Turin, Turin, Italy
| | - Erica Silvestris
- Gynecologic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II," Bari, Italy
| | - Andrea Roberto Carosso
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant'Anna Hospital, University of Turin, Turin, Italy
| | - Alessandro Ruffa
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant'Anna Hospital, University of Turin, Turin, Italy
| | - Bernadette Evangelisti
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant'Anna Hospital, University of Turin, Turin, Italy
| | - Gianluca Gennarelli
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant'Anna Hospital, University of Turin, Turin, Italy
| | - Gennaro Cormio
- Gynecologic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II," Bari, Italy; Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Vera Loizzi
- Gynecologic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II," Bari, Italy; Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Alessandro Rolfo
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Chiara Benedetto
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant'Anna Hospital, University of Turin, Turin, Italy
| | - Alberto Revelli
- Gynecology and Obstetrics 2U, Department of Surgical Sciences, S. Anna Hospital, University of Turin, Turin, Italy
| |
Collapse
|
23
|
Armstrong R, Marks NJ, Geary TG, Harrington J, Selzer PM, Maule AG. Wnt/β-catenin signalling underpins juvenile Fasciola hepatica growth and development. PLoS Pathog 2025; 21:e1012562. [PMID: 39919127 PMCID: PMC11805424 DOI: 10.1371/journal.ppat.1012562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025] Open
Abstract
Infection by the liver fluke, Fasciola hepatica, places a substantial burden on the global agri-food industry and poses a significant threat to human health in endemic regions. Widespread resistance to a limited arsenal of chemotherapeutics, including the frontline flukicide triclabendazole (TCBZ), renders F. hepatica control unsustainable and accentuates the need for novel therapeutic target discovery. A key facet of F. hepatica biology is a population of specialised stem cells which drive growth and development - their dysregulation is hypothesised to represent an appealing avenue for control. The exploitation of this system as a therapeutic target is impeded by a lack of understanding of the molecular mechanisms underpinning F. hepatica growth and development. Wnt signalling pathways govern a myriad of stem cell processes during embryogenesis and drive tumorigenesis in adult tissues in animals. Here, we identify five putative Wnt ligands and five Frizzled receptors in liver fluke transcriptomic datasets and find that Wnt/β-catenin signalling is most active in juveniles, the most pathogenic life stage. FISH-mediated transcript localisation revealed partitioning of the five Wnt ligands, with each displaying a distinct expression pattern, consistent with each Wnt regulating the development of different cell/tissue types. The silencing of each individual Wnt or Frizzled gene yielded significant reductions in juvenile worm growth and, in select cases, blunted the proliferation of neoblast-like cells. Notably, silencing FhCTNNB1, the key effector of the Wnt/β-catenin signal cascade led to aberrant development of the neuromuscular system which ultimately proved lethal - the first report of a lethal RNAi-induced phenotype in F. hepatica. The absence of any discernible phenotypes following the silencing of the inhibitory Wnt/β-catenin destruction complex components is consistent with low destruction complex activity in rapidly developing juvenile worms, corroborates transcriptomic expression profiles and underscores the importance of Wnt signalling as a key molecular driver of growth and development in early-stage juvenile fluke. The putative pharmacological inhibition of Wnt/β-catenin signalling using commercially available inhibitors phenocopied RNAi results and provides impetus for drug repurposing. Taken together, these data functionally and chemically validate the targeting of Wnt signalling as a novel strategy to undermine the pathogenicity of juvenile F. hepatica.
Collapse
Affiliation(s)
- Rebecca Armstrong
- Understanding Health and Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Nikki J. Marks
- Understanding Health and Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Timothy G. Geary
- Understanding Health and Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | - John Harrington
- Parasitology, Boehringer Ingelheim Animal Health, Duluth, Georgia, United States of America
| | - Paul M. Selzer
- Parasitology, Boehringer Ingelheim Vetmedica GmbH, Ingelheim am Rhein, Germany
| | - Aaron G. Maule
- Understanding Health and Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| |
Collapse
|
24
|
Zan GX, Qu HZ, Meng J, Wang XF, Yan HC, Wang XQ, Zhou JY. Matrine disturbs the eimeria necatrix-induced loop of tuft cell-intestinal stem cell-goblet cell by inactivating IL-13/JAK2/STAT3 signaling. Poult Sci 2025; 104:104786. [PMID: 39798285 PMCID: PMC11954915 DOI: 10.1016/j.psj.2025.104786] [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: 08/30/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/15/2025] Open
Abstract
As sensors in the gut, tuft cells integrate a complex array of luminal signals to regulate the differentiation fate of intestinal stem cells (ISCs), which trigger a loop of tuft cell-ISC-goblet cell after parasitic infection. As a plant-derived alkaloid, Matrine plays a prominent role for standardizing ISC functions in Eimeria necatrix (EN)-exposed chicks. In this study, we investigated the modulation effects of Matrine on the specific intestinal epithelial cell loop in EN-exposed chicks in vivo and intestinal organoids (IOs) ex vivo. The results showed that EN infection resulted in swelling and hemorrhage of the jejunum, accompanied by the increase in levels of sIgA and inflammatory cytokines (IL-6, IL-1β, and TNF-α). And these inflammatory symptoms were effectively relieved by Matrine intervention. Concurrently, Matrine resisted the EN-induced increase in tuft cell numbers and levels of crucial pro-inflammatory factors (IL-25 and IL-13), while also reversing the differentiation of secretory cell progenitors into goblet cells. Importantly, Matrine impeded the upregulation of the inflammatory signaling pathway JAK2/STAT3 in EN-infected chicks and IOs. Conversely, exogenous supplementation of IL-13 or activation of STAT3 via Colivelin eliminated the standardization of the tuft cell-ISC-goblet cell loop by Matrine. Overall, our findings suggested that Matrine intercepted the tuft cell-ISC-goblet cell loop by reinstating IL-13/JAK2/STAT3 signaling after EN infection.
Collapse
Affiliation(s)
- Geng-Xiu Zan
- State Key Laboratory of Swine and Poultry Breeding Industry/College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, 510642, China
| | - Hao-Zhan Qu
- State Key Laboratory of Swine and Poultry Breeding Industry/College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, 510642, China
| | - Jia Meng
- State Key Laboratory of Swine and Poultry Breeding Industry/College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, 510642, China
| | - Xiao-Fan Wang
- State Key Laboratory of Swine and Poultry Breeding Industry/College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, 510642, China
| | - Hui-Chao Yan
- State Key Laboratory of Swine and Poultry Breeding Industry/College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, 510642, China
| | - Xiu-Qi Wang
- State Key Laboratory of Swine and Poultry Breeding Industry/College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, 510642, China
| | - Jia-Yi Zhou
- State Key Laboratory of Swine and Poultry Breeding Industry/College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou, 510642, China.
| |
Collapse
|
25
|
Good HJ, Larsen F, Shin AE, Zhang L, Derouet M, Meriwether D, Worthley D, Reddy ST, Wang TC, Asfaha S. Prostaglandin E 2 and Akt Promote Stemness in Apc Mutant Dclk1+ Cells to Give Rise to Colitis-associated Cancer. Cell Mol Gastroenterol Hepatol 2025; 19:101469. [PMID: 39884575 PMCID: PMC11999635 DOI: 10.1016/j.jcmgh.2025.101469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND & AIMS Loss of the tumor suppressor gene Apc in Lgr5+ intestinal stem cells results in aberrant Wnt signaling and colonic tumorigenesis. In the setting of injury, however, we and others have also shown that non-stem cells can give rise to colonic tumors. The mechanism by which inflammation leads to cellular plasticity and cancer, however, remains largely unknown. METHODS RNA expression analysis of Wnt, COX, and Akt signaling was assessed in patients with quiescent or active ulcerative colitis (UC) and patients with UC-associated neoplasia using available datasets. The role of COX signaling in colonic tumorigenesis was examined using epithelial and doublecortin-like kinase 1 (Dclk1)+ cell-specific conditional COX-1 knockout mice and pharmacologic treatment with different nonsteroidal anti-inflammatory drugs. RESULTS In this study, we show that prostaglandins and phospho-Akt are key inflammatory mediators that promote stemness in Apc mutant Dclk1+ cells that give rise to colorectal cancer. Moreover, prostaglandin E2 (PGE2) and Akt are increased in colitis in both mice and humans, leading to inflammation-associated dysplasia upon activation of Wnt signaling. Importantly, inhibition of epithelial-derived COX-1 by aspirin or conditional knockout in Dclk1+ cells reduced PGE2 levels and prevented the development of inflammation-associated colorectal cancer. CONCLUSIONS Our data shows that epithelial and Dclk1+ cell-derived COX-1 plays an important role in inflammation-associated tumorigenesis. Importantly, low-dose aspirin was effective in chemo-prevention through inhibition of COX-1 that reduced colitis-associated cancer.
Collapse
Affiliation(s)
- Hayley J Good
- Department of Medicine, University of Western Ontario, London, Ontario, Canada; Verspeeten Family Cancer Centre, London Health Sciences Centre, London, Ontario, Canada
| | - Frederikke Larsen
- Department of Medicine, University of Western Ontario, London, Ontario, Canada; Verspeeten Family Cancer Centre, London Health Sciences Centre, London, Ontario, Canada
| | - Alice E Shin
- Department of Medicine, University of Western Ontario, London, Ontario, Canada; Verspeeten Family Cancer Centre, London Health Sciences Centre, London, Ontario, Canada
| | - Liyue Zhang
- Department of Medicine, University of Western Ontario, London, Ontario, Canada; Verspeeten Family Cancer Centre, London Health Sciences Centre, London, Ontario, Canada
| | - Mathieu Derouet
- Department of Medicine, University of Western Ontario, London, Ontario, Canada; Verspeeten Family Cancer Centre, London Health Sciences Centre, London, Ontario, Canada
| | - David Meriwether
- Department of Medicine, Division of Cardiology, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Daniel Worthley
- South Australian Health Medical Research Institute, North Terrace Adelaide, Australia
| | - Srinivasa T Reddy
- Department of Medicine, Division of Cardiology, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, New York
| | - Samuel Asfaha
- Department of Medicine, University of Western Ontario, London, Ontario, Canada; Verspeeten Family Cancer Centre, London Health Sciences Centre, London, Ontario, Canada.
| |
Collapse
|
26
|
Zan G, He H, Wang X, Zhou J, Wang X, Yan H. Morin Reactivates Nrf2 by Targeting Inhibition of Keap1 to Alleviate Deoxynivalenol-Induced Intestinal Oxidative Damage. Int J Mol Sci 2025; 26:1086. [PMID: 39940854 PMCID: PMC11817132 DOI: 10.3390/ijms26031086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
As a prevalent mycotoxin found in cereal foods and feed, deoxynivalenol (DON) disrupts the orderly regeneration of intestinal epithelial tissue by interfering with the intracellular antioxidant defense system. However, the potential of mulberry leaf-derived Morin, a natural flavonoid active substance with clearing reactive oxygen species (ROS), to mitigate DON-induced intestinal oxidative damage remains unclear. Our investigation demonstrates that Morin effectively reverses the decline in growth performance and repairs damaged jejunal structures and barrier function under DON exposure. Furthermore, the proliferation and differentiation of intestinal stem cells (ISCs) is enhanced significantly after Morin intervention. Importantly, Morin increases the levels of total antioxidant capacity (T-AOC), superoxide dismutase (SOD), and glutathione peroxidase (GSH-PX) in the serum and jejunal tissue, while reducing the accumulation of ROS and malondialdehyde (MDA). Molecular interaction analysis further confirms that Morin targets inhibition of Keap1 to activate the Nrf2-mediated antioxidant system. In summary, our results suggest that Morin alleviates the oxidative damage induced by DON by regulating the Keap1/Nrf2 pathway, thereby restoring the proliferation and differentiation activity of ISC, which provides new insights into Morin mitigating DON damage.
Collapse
Affiliation(s)
| | | | | | | | | | - Huichao Yan
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (G.Z.); (H.H.); (X.W.); (J.Z.); (X.W.)
| |
Collapse
|
27
|
Wang H, He X, Zhang M, Fan N, Yang Z, Shen T, Guo J, Song Y, Cao G, Liu Y, Li X, Nashun B. Development of Sheep Intestinal Organoids for Studying Deoxynivalenol-Induced Toxicity. Int J Mol Sci 2025; 26:955. [PMID: 39940725 PMCID: PMC11816529 DOI: 10.3390/ijms26030955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Sheep are an important livestock species whose gastrointestinal tract is essential for overall health. Feed contaminants such as bacterial toxins and mycotoxins severely damage the sheep intestine, yet the mechanisms remain mostly elusive partially due to the lack of physiologically relevant in vitro models. Here, we investigated molecular mechanisms underlying deoxynivalenol (DON)-induced toxicity by developing intestinal organoids from isolated intestinal crypts of Hu sheep. The organoids had a central lumen and monolayer epithelium, and could be continuously passaged, cryopreserved, and resuscitated. Histological and transcriptomic analysis showed that the intestinal organoids recapitulate the cell lineages and gene expression characteristics of the original intestinal tissues. Statistical analysis indicated that DON exposure significantly inhibited organoid formation efficiency, as well as the proliferation and activity of intestinal organoid cells. RNA-seq and Western blotting analysis further revealed that DON exposure induces intestinal toxicity by inhibiting the PI3K/AKT/GSK3β/β-catenin signaling pathway. Our study provides a novel example of organoid application in toxicity studies and reveals the signaling pathway involved in DON-induced toxicity in sheep, which is of great significance for improving mitigation strategies for DON.
Collapse
Affiliation(s)
- Hongyu Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
| | - Xige He
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
| | - Miaomiao Zhang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
| | - Na Fan
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
| | - Zongxuan Yang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
| | - Ting Shen
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
| | - Jiaojiao Guo
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
| | - Yongli Song
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
| | - Guifang Cao
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
- Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic Animals, Hohhot 011517, China
| | - Yongbin Liu
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
| | - Xihe Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
- Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic Animals, Hohhot 011517, China
| | - Buhe Nashun
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China; (H.W.); (X.H.); (M.Z.); (N.F.); (Z.Y.); (T.S.); (J.G.); (Y.S.); (G.C.); (Y.L.)
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
| |
Collapse
|
28
|
Jang JH, Jung J, Kang HG, Kim W, Kim WJ, Lee H, Cho JY, Hong R, Kim JW, Chung JY, Chun KH, Kim SJ. Kindlin-1 promotes gastric cancer cell motility through the Wnt/β-catenin signaling pathway. Sci Rep 2025; 15:2481. [PMID: 39833319 PMCID: PMC11756408 DOI: 10.1038/s41598-025-86220-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Despite advances in gastric cancer diagnosis and treatment, its prognosis remains poor owing to aggressive tumor progression and metastasis. As understanding the relevant molecular mechanisms is essential to effectively improve patient outcomes, we elucidated the role of Kindlin-1 in gastric cancer progression and metastasis. Kindlin-1 expression was analyzed in 359 gastric cancer tissue samples provided by Kangnam Sacred Heart Hospital and publicly available GSE datasets. Kindlin-1 showed significantly higher expression in gastric cancer tissues than that in normal tissues, and high Kindlin-1 expression was associated with poor prognosis. Further, the mRNA and protein expression of Kindlin-1 were high in gastric cancer cell lines, where they were associated with increased proliferation, migration, and invasion. Our findings demonstrated that Kindlin-1 regulated epithelial-mesenchymal transition-related genes through interaction with activated Wnt/β-catenin signaling. Notably, Kindlin-1 enhanced β-catenin expression and promoted its nuclear translocation from the cytoplasm, increasing TCF4 transcriptional activity and inducing gastric cancer progression and metastasis. Overall, these findings demonstrate that Kindlin-1 is upregulated in gastric cancer and activates Wnt/β-catenin signaling to promote cell proliferation and motility.
Collapse
Affiliation(s)
- Jun-Ho Jang
- Department of Integrative Biological Sciences and BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju, 61452, Republic of Korea
| | - Jiyoon Jung
- Department of Pathology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, 07441, Republic of Korea
| | - Hyeon-Gu Kang
- Department of Integrative Biological Sciences and BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju, 61452, Republic of Korea
- Institute of Well-Aging Medicare and Chosun University G-LAMP Project Group, Chosun University, Gwangju, 61452, Republic of Korea
| | - Woong Kim
- Institute of Well-Aging Medicare and Chosun University G-LAMP Project Group, Chosun University, Gwangju, 61452, Republic of Korea
| | - Won-Jin Kim
- Department of Integrative Biological Sciences and BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju, 61452, Republic of Korea
- Institute of Well-Aging Medicare and Chosun University G-LAMP Project Group, Chosun University, Gwangju, 61452, Republic of Korea
| | - Hana Lee
- Department of Integrative Biological Sciences and BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju, 61452, Republic of Korea
- Institute of Well-Aging Medicare and Chosun University G-LAMP Project Group, Chosun University, Gwangju, 61452, Republic of Korea
| | - Ju Yeon Cho
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, 61452, Republic of Korea
| | - Ran Hong
- Department of Pathology, College of Medicine, Chosun University, Gwangju, 61452, Republic of Korea
| | - Jeong Won Kim
- Department of Pathology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, 07441, Republic of Korea
| | - Joon-Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kyung-Hee Chun
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Seok-Jun Kim
- Department of Integrative Biological Sciences and BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju, 61452, Republic of Korea.
- Institute of Well-Aging Medicare and Chosun University G-LAMP Project Group, Chosun University, Gwangju, 61452, Republic of Korea.
- Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
29
|
Filippi A, Deculescu-Ioniță T, Hudiță A, Baldasici O, Gălățeanu B, Mocanu MM. Molecular Mechanisms of Dietary Compounds in Cancer Stem Cells from Solid Tumors: Insights into Colorectal, Breast, and Prostate Cancer. Int J Mol Sci 2025; 26:631. [PMID: 39859345 PMCID: PMC11766403 DOI: 10.3390/ijms26020631] [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: 11/21/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Cancer stem cells (CSC) are known to be the main source of tumor relapse, metastasis, or multidrug resistance and the mechanisms to counteract or eradicate them and their activity remain elusive. There are different hypotheses that claim that the origin of CSC might be in regular stem cells (SC) and, due to accumulation of mutations, these normal cells become malignant, or the source of CSC might be in any malignant cell that, under certain environmental circumstances, acquires all the qualities to become CSC. Multiple studies indicate that lifestyle and diet might represent a source of wellbeing that can prevent and ameliorate the malignant phenotype of CSC. In this review, after a brief introduction to SC and CSC, we analyze the effects of phenolic and non-phenolic dietary compounds and we highlight the molecular mechanisms that are shown to link diets to CSC activation in colon, breast, and prostate cancer. We focus the analysis on specific markers such as sphere formation, CD surface markers, epithelial-mesenchymal transition (EMT), Oct4, Nanog, Sox2, and aldehyde dehydrogenase 1 (ALDH1) and on the major signaling pathways such as PI3K/Akt/mTOR, NF-κB, Notch, Hedgehog, and Wnt/β-catenin in CSC. In conclusion, a better understanding of how bioactive compounds in our diets influence the dynamics of CSC can raise valuable awareness towards reducing cancer risk.
Collapse
Affiliation(s)
- Alexandru Filippi
- Department of Biochemistry and Biophysics, “Carol Davila” University of Medicine and Pharmacy of Bucharest, 050474 Bucharest, Romania;
| | - Teodora Deculescu-Ioniță
- Department of Pharmacognosy, Phytochemistry and Phytotherapy, “Carol Davila” University of Medicine and Pharmacy of Bucharest, 050474 Bucharest, Romania;
| | - Ariana Hudiță
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania; (A.H.); (B.G.)
| | - Oana Baldasici
- Department of Genetics, Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuță”, 400015 Cluj-Napoca, Romania;
| | - Bianca Gălățeanu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania; (A.H.); (B.G.)
| | - Maria-Magdalena Mocanu
- Department of Biochemistry and Biophysics, “Carol Davila” University of Medicine and Pharmacy of Bucharest, 050474 Bucharest, Romania;
| |
Collapse
|
30
|
Yan P, Guo Y, Muhammad S, Zhu J, Liu Y, Liu C. The effects of the Wnt/β-catenin signaling pathway on the in vitro differentiation of rat BMSCs into leydig cells. Sci Rep 2025; 15:1177. [PMID: 39775149 PMCID: PMC11707357 DOI: 10.1038/s41598-025-85674-z] [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: 10/08/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
Late-onset hypogonadism (LOH) refers to sexual and non-sexual symptoms in men caused by age-related decreases in circulating testosterone. Leydig cells (LCs) transplantation is considered to be one of a viable approach for LOH therapy, but the limited source of LCs limits the application of this approach. The aim of this study was to induce the directed differentiation of rat bone marrow mesenchymal stem cells (BMSCs) into LCs in vitro, and explore the potential involvement of Wnt/β-catenin signaling pathway in the differentiation process. BMSCs were extracted from rats and characterized by flow cytometry for positive rates of mesenchymal stem cell markers CD29, CD44, CD90, and the hematopoietic marker CD45. BMSCs were divided into three groups: Control, Wnt agonist (CHIR-99021), and Wnt inhibitor (LGK-974), each incubated for 14 days. ELISA and RT-qPCR were used to verify the protein and mRNA expression of β-catenin, LRP5 and TCF, the key factors in Wnt/β-catenin signaling pathway. The average fluorescence intensity of 3β-hydroxysteroid dehydrogenase (3β-HSD) on the surface of LCs was detected by immunofluorescence (IF) assay. The content of testosterone secreted in cell culture medium was detected by ELISA. The results of flow cytometry indicated that we successfully extracted and cultured BMSCs. Moreover, post 14 days of incubation, the changes of β-catenin, LRP5 and TCF, at the protein and mRNA level demonstrate successful intervention in the activation and inhibition of the intracellular Wnt/β-catenin signaling pathway. Compared with the control group, the LCs surface marker 3β-HSD expression intensity in the CHIR-99,021 group was significantly increased by 69% (p < 0.01), while significantly decreased by 59% in LGK-974 group (p < 0.01). The ELISA results indicated a higher testosterone concentration in the CHIR-99,021 group (359.58 ± 17.46 pg/mL) than in the control (225.31 ± 15.42 pg/mL) and LGK-974 groups (183.67 ± 4.47 pg/mL), and the difference was statistically significant (p < 0.05). This study successfully demonstrates the directed differentiation of BMSCs into LCs under the action of inducers. We verified that the Wnt/β-catenin signaling pathway is involved in this differentiation process. The idea proposed in our study for efficiently inducing differentiation of BMSCs into LC in vitro, may provide a safe and sustainable LC source for developing clinically feasible cell transplantation-based LOH therapies.
Collapse
Affiliation(s)
- Pengyu Yan
- First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, China
- Department of Urology, First Hospital of Shanxi Medical University, No. 85, Jiefang South Road, Taiyuan, 030001, China
| | - Yaxiong Guo
- First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, China
- Department of Urology, First Hospital of Shanxi Medical University, No. 85, Jiefang South Road, Taiyuan, 030001, China
| | - Shoaib Muhammad
- First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, China
| | - Jinxiong Zhu
- Department of Urology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Yuxiang Liu
- Department of Nephrology, Shanxi Provincial People 's Hospital, No. 29, Shuangta Street, Taiyuan, 030012, China.
| | - Chun Liu
- Department of Urology, First Hospital of Shanxi Medical University, No. 85, Jiefang South Road, Taiyuan, 030001, China.
| |
Collapse
|
31
|
Uno K, Uchino T, Suzuki T, Sayama Y, Edo N, Uno-Eder K, Morita K, Ishikawa T, Koizumi M, Honda H, Katagiri H, Tsukamoto K. Rspo3-mediated metabolic liver zonation regulates systemic glucose metabolism and body mass in mice. PLoS Biol 2025; 23:e3002955. [PMID: 39854351 PMCID: PMC11759367 DOI: 10.1371/journal.pbio.3002955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 11/27/2024] [Indexed: 01/26/2025] Open
Abstract
The unique architecture of the liver consists of hepatic lobules, dividing the hepatic features of metabolism into 2 distinct zones, namely the pericentral and periportal zones, the spatial characteristics of which are broadly defined as metabolic zonation. R-spondin3 (Rspo3), a bioactive protein promoting the Wnt signaling pathway, regulates metabolic features especially around hepatic central veins. However, the functional impact of hepatic metabolic zonation, regulated by the Rspo3/Wnt signaling pathway, on whole-body metabolism homeostasis remains poorly understood. In this study, we analyze the local functions of Rspo3 in the liver and the remote actions of hepatic Rspo3 on other organs of the body by using murine models. Rspo3 expression analysis shows that Rspo3 expression patterns are spatiotemporally controlled in the murine liver such that it locates in the pericentral zones and converges after feeding, and the dynamics of these processes are disturbed in obesity. We find that viral-mediated induction of Rspo3 in hepatic tissue of obesity improves insulin resistance and prevents body weight gain by restoring attenuated organ insulin sensitivities, reducing adipose tissue enlargement and reversing overstimulated adaptive thermogenesis. Denervation of the hepatic vagus suppresses these remote effects, derived from hepatic Rspo3 induction, toward adipose tissues and skeletal muscle, suggesting that signals are transduced via the neuronal communication consisting of afferent vagal and efferent sympathetic nerves. Furthermore, the non-neuronal inter-organ communication up-regulating muscle lipid utilization is partially responsible for the ameliorations of both fatty liver development and reduced skeletal muscle quality in obesity. In contrast, hepatic Rspo3 suppression through Cre-LoxP-mediated recombination system exacerbates diabetes due to glucose intolerance and insulin resistance, promotes fatty liver development and decreases skeletal muscle quality, resulting in obesity. Taken together, our study results reveal that modulation of hepatic Rspo3 contributes to maintaining systemic glucose metabolism and body composition via a newly identified inter-organ communication mechanism.
Collapse
Affiliation(s)
- Kenji Uno
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Takuya Uchino
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Takashi Suzuki
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Yohei Sayama
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Naoki Edo
- Teikyo Academic Research Center, Tokyo, Japan
| | | | - Koji Morita
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Toshio Ishikawa
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Miho Koizumi
- Field of Human Disease Models, Tokyo Women’s Medical University, Tokyo, Japan
| | - Hiroaki Honda
- Field of Human Disease Models, Tokyo Women’s Medical University, Tokyo, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuhisa Tsukamoto
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
32
|
Wu X, Wang S, Pan Y, Li M, Song M, Zhang H, Deng M, Yang X, Xu J, Zhang S, Zhang J, Wang F, Plikus MV, Lv C, Yu L, Yu Z. m 6A Reader PRRC2A Promotes Colorectal Cancer Progression via CK1ε-Mediated Activation of WNT and YAP Signaling Pathways. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406935. [PMID: 39582289 PMCID: PMC11744581 DOI: 10.1002/advs.202406935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/12/2024] [Indexed: 11/26/2024]
Abstract
Colorectal cancer (CRC) is the third most common cancer type and the second highest mortality rate among cancers. However, the mechanisms underlying CRC progression remain to be fully understood. In this work, a recently identified m6A-modified RNA reader protein Proline-rich Coiled-coil 2a (PRRC2A) is markedly upregulated in CRC, and intestinal epithelium-specific deletion of Prrc2a significantly suppressed tumor cell growth, stemness, and migratory capacity, while its overexpression promoted these behaviors. Through multiomics analysis, PRRC2A directly targeted CSNK1E (encoding CK1ε), maintaining its RNA stability in an m6A-dependent manner, and that elevated CK1ε can concomitantly result in activation of the WNT and YAP signaling pathways. Interestingly, PRRC2A is directly regulated by the transcription factor ATF1 in its promoter. In summary, the work reveals a novel mechanism by which m6A reader PRRC2A promotes colorectal cancer progression via CK1ε and aberrant upregulation of WNT and YAP signaling. Therefore, PRRC2A and CK1ε can be potential therapeutic targets for treating CRC.
Collapse
Affiliation(s)
- Xi Wu
- The First Affiliated Hospital of Zhengzhou UniversityTianjian Laboratory of Advanced Biomedical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450052China
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Shiyang Wang
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Yuwei Pan
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Mengzhen Li
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Manyu Song
- Key Laboratory of Precision Nutrition and Food QualityMinistry of EducationDepartment of Nutrition and HealthChina Agricultural UniversityBeijing100193China
| | - Hanfu Zhang
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Min Deng
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Xu Yang
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Jiuzhi Xu
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Shuo Zhang
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Jinhua Zhang
- The college of Life Science and BioengineeringBeijing Jiaotong UniversityBeijing100044China
| | - Fengchao Wang
- National Institute of Biological ScienceBeijing102206China
| | - Maksim V. Plikus
- Department of Developmental and Cell BiologySue and Bill Gross Stem Cell Research CenterCenter for Complex Biological SystemsUniversity of CaliforniaIrvineCA92697USA
| | - Cong Lv
- Key Laboratory of Precision Nutrition and Food QualityMinistry of EducationDepartment of Nutrition and HealthChina Agricultural UniversityBeijing100193China
| | - Lu Yu
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Zhengquan Yu
- The First Affiliated Hospital of Zhengzhou UniversityTianjian Laboratory of Advanced Biomedical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450052China
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| |
Collapse
|
33
|
Kumari R, Ghava D, Rathod R, Panda AK, Kumar S, Behera SK. Molecular Dynamics of Adenomatous Polyposis Coli (APC) Protein and Its Inhibitors: A Special Insight to Colorectal Cancer. Crit Rev Oncog 2025; 30:91-105. [PMID: 39819437 DOI: 10.1615/critrevoncog.v30.i1.110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Colorectal cancer (CRC) initiates in colon or rectum is named as colon or rectal cancer, based on the site of inception. Various genetic alterations responsible for CRC include several signaling pathways. The Wingless/Wnt signaling pathway is the vital pathway which involved in the cancer pathogenesis. The hallmark of human CRC is adenomatous polyposis coli (APC), a negative regulator of the Wnt pathway. Mutations in the APC gene is a critical event in the development of human CRC which may lead to overexpression and stabilization of β-catenin that enters into the nucleus and helps in cancer cell proliferation. Significant obstacles to the therapeutic intervention of the Wnt signaling system still exist, despite promising approaches for the development of anti-cancer medicines targeting this route. The advent of computational techniques for cancer diagnosis, prognosis, and drug development has spurred the researchers to explore CRC at an early stage. This report had unzipped the importance of APC in Wnt signaling pathway associated with current advances and challenges in drug discovery for CRC. A combinatorial computational approach identified the potential anti-cancerous drug among XL888, 5-bromouracil, 5-fluorouracil, and Ganetespib against APC which is often treated as gatekeeper of CRC. This in silico investigation revealed Ganetespib as a potential anti-cancerous drug against APC for CRC therapeutics, which will be an alternative to chemotherapy. In vitro and in vivo studies are needed further to confirm the efficiency and evaluate potency of Ganetespib against the target.
Collapse
Affiliation(s)
- Rina Kumari
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, India 382055
| | - Dilip Ghava
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, India 382055
| | - Rajeshwari Rathod
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, India 382055
| | | | - Sunil Kumar
- ICAR-Indian Agricultural Statistics Research Institute, New Delhi, India
| | - Santosh Kumar Behera
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, India -382055, Gujarat, India
| |
Collapse
|
34
|
Zheng B, Zhang W, Xie S, Han Y, Liu G, Liu Y, Gao M, Wang S, Liu Q. Selinexor as a Therapeutic Target: Advances in Non-small Cell and Small Cell Lung Cancer Treatment Strategies. Recent Pat Anticancer Drug Discov 2025; 20:274-284. [PMID: 39473203 DOI: 10.2174/0115748928322627241016120142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/21/2024] [Accepted: 09/04/2024] [Indexed: 04/24/2025]
Abstract
Selinexor treats lung cancer, particularly non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). This review summarizes the prevalence and types of lung cancer and emphasizes the challenges associated with current treatments like resistance and limited effectiveness. Selinexor is a selective inhibitor of nuclear export (SINE) that has emerged as a potential therapy that targets the nuclear export of tumor suppressor proteins. The mechanisms of selinexor, its potential in combination therapies, and challenges like side effects and drug resistance are explained in this review. Key findings highlight the effectiveness of selinexor in preclinical studies, particularly against KRAS-mutant NSCLC and in combination with chemotherapy for SCLC. The review concludes with a discussion of future directions and underscores the potential of selinexor to improve the treatment strategies for lung cancer.
Collapse
Affiliation(s)
- Bosheng Zheng
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wenqi Zhang
- Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shaonan Xie
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yaqing Han
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guangjie Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yanjie Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Maogang Gao
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shize Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qingyi Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
35
|
Wang D, Wu N, Li P, Zhang X, Xie W, Li S, Wang D, Kuang Y, Chen S, Liu Y. Eicosapentaenoic acid enhances intestinal stem cell-mediated colonic epithelial regeneration by activating the LSD1-WNT signaling pathway. J Adv Res 2024:S2090-1232(24)00628-3. [PMID: 39743214 DOI: 10.1016/j.jare.2024.12.050] [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: 08/11/2024] [Revised: 12/26/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025] Open
Abstract
INTRODUCTION Inflammatory bowel disease (IBD) is often associated with impaired proliferation and differentiation of intestinal stem cells (ISCs). Eicosapentaenoic acid (EPA), which is predominantly found in fish oil, has been recognized for its intestinal health benefits, although the potential mechanisms are not well understood. OBJECTIVES This study aimed to investigate the regulatory role and mechanism of EPA in colonic epithelial regeneration, specifically from the perspective of ISCs. METHODS Wild-type mice whose diet was supplemented with 5% EPA-enriched fish oil were subjected to dextran sulfate sodium (DSS) to induce colitis. We utilized intestinal organoids, ISC-specific lysine-specific demethylase 1 (LSD1) knockout mice, and WNT inhibitor-treated mice to explore how EPA influences ISC proliferation and differentiation. ISC proliferation, differentiation and apoptosis were assessed using tdTomato and propidium iodide tracer testing, histological analyses, and immunofluorescence staining. RESULTS EPA treatment significantly mitigated the symptoms of DSS-induced acute colitis, as evidenced by lower body weight loss and decreased disease activity index, histological scores and proinflammatory cytokine levels. Additionally, EPA increased the numbers of proliferative cells, absorptive cells, goblet cells, and enteroendocrine cells, which enhanced the regeneration of intestinal epithelium. Pretreatment with EPA increased ISC proliferation and differentiation, and protected against TNF-α-induced cell death in intestinal organoids. Mechanistically, EPA upregulated G protein-coupled receptor 120 (GPR120) to induce LSD1 expression, which facilitated ISC proliferation and differentiation in organoids. ISC-specific ablation of LSD1 negated the protective effect of EPA on DSS-induced colitis in mice. Moreover, EPA administration activated the WNT signaling pathway downstream of LSD1 in ISCs, while inhibiting WNT signaling abolished the beneficial effects of EPA. CONCLUSIONS These findings demonstrate that EPA promotes ISC proliferation and differentiation, thereby enhancing colonic epithelial regeneration through the activation of LSD1-WNT signaling. Consequently, dietary supplementation with EPA represents a promising alternative therapeutic strategy for managing IBD.
Collapse
Affiliation(s)
- Dan Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
| | - Nianbang Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
| | - Pei Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
| | - Xiaojuan Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
| | - Wenshuai Xie
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
| | - Shunkang Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
| | - Ding Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yanling Kuang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
| | - Shaokui Chen
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China.
| |
Collapse
|
36
|
Alimogullari E, Kartal B, Demir H, Elci MP. Protective effects of adipose-derived stem cells against testicular injury induced after ischemia-reperfusion by regulating autophagy. Histochem Cell Biol 2024; 163:18. [PMID: 39709318 DOI: 10.1007/s00418-024-02347-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2024] [Indexed: 12/23/2024]
Abstract
The damaged organ may experience severe pathological alterations as a result of tissue ischemia-reperfusion (I/R). The study of stem cell-based repair therapies is actively being conducted, and the outcomes and therapeutic potential of these cells are both promising. Autophagy checks protein homeostasis by breaking down huge damaged proteins or organelles. The study's objective was to assess how ADSCs impact the autophagic process after testicular ischemia/reperfusion. In our investigation, 30 male rats were divided into five groups: control, ADSC, ischemia, I/R, and I/R + ADSC (n = 6). Hematoxylin-eosin (HE) was used to stain the testes, and histological changes were assessed. The immunoexpression of androgen receptor (AR), Beclin1, protein light chain 3B (LC3B), and p62 were examined. The seminiferous epithelium in the testis from the ischemia and I/R groups revealed significant degeneration with disorganized morphology, damaged spermatogenic cells, and interstitial space congestion, according to HE stain results. Johnsen's score were significantly better in I/R + ADSC group than in ischemia and I/R groups. We demonstrated that in rat testes from the I/R groups, immunostaining of Beclin 1 (p = 0.042) and LC3B (p = 0.011) were raised, and p62 (p = 0.047) and AR (p = 0.049) were decreased. Ischemia and I/R promoted testicular autophagy, therefore we can conclude that ADSCs prevent excessive autophagy. Additionally, these results show that the use of ADSCs cures testicular injury and dysfunction associated with I/R injury in rats even a little.
Collapse
Affiliation(s)
- Ebru Alimogullari
- Medical Faculty, Department of Histology and Embryology, Ankara Yıldırım Beyazıt University, Ankara, Turkey.
| | - Bahar Kartal
- Medical Faculty, Department of Histology and Embryology, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Hazal Demir
- Medical Faculty, Department of Histology and Embryology, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Mualla Pınar Elci
- Stem Cell Laboratory, University of Health Sciences Gulhane Health Sciences Institute, Ankara, Turkey
| |
Collapse
|
37
|
Kita K, Morkos C, Nolan K. Maintenance of stem cell self-renewal by sex chromosomal zinc-finger transcription factors. World J Methodol 2024; 14:97664. [PMID: 39712568 PMCID: PMC11287546 DOI: 10.5662/wjm.v14.i4.97664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 07/26/2024] Open
Abstract
In this Editorial review, we would like to focus on a very recent discovery showing the global autosomal gene regulation by Y- and inactivated X-chromosomal transcription factors, zinc finger gene on the Y chromosome (ZFY) and zinc finger protein X-linked (ZFX). ZFX and ZFY are both zinc-finger proteins that encode general transcription factors abundant in hematopoietic and embryonic stem cells. Although both proteins are homologs, interestingly, the regulation of self-renewal by these transcriptional factors is almost exclusive to ZFX. This fact implies that there are some differential roles between ZFX and ZFY in regulating the maintenance of self-renewal activity in stem cells. Besides the maintenance of stemness, ZFX overexpression or mutations may be linked to certain cancers. Although cancers and stem cells are double-edged swords, there is no study showing the link between ZFX activity and the telomere. Thus, stemness or cancers with ZFX may be linked to other molecules, such as Oct4, Sox2, Klf4, and others. Based on very recent studies and a few lines of evidence in the past decade, it appears that the ZFX is linked to the canonical Wnt signaling, which is one possible mechanism to explain the role of ZFX in the self-renewal of stem cells.
Collapse
Affiliation(s)
- Katsuhiro Kita
- Department of Biology, St. Francis College, Brooklyn, NY 11201, United States
| | - Celine Morkos
- Department of Biology, St. Francis College, Brooklyn, NY 11201, United States
| | - Kathleen Nolan
- Department of Biology, St. Francis College, Brooklyn, NY 11201, United States
| |
Collapse
|
38
|
Koval A, Boudou C, Katanaev VL. Challenging Reported Frizzled-Targeting Compounds in Selective Assays Reveals Lack of Functional Inhibition and Claimed Profiles. ACS Pharmacol Transl Sci 2024; 7:4144-4154. [PMID: 39698282 PMCID: PMC11650735 DOI: 10.1021/acsptsci.4c00570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
Selective inhibitors of Frizzled (FZD) GPCRs are highly sought after as potentially highly efficacious and safe treatments for cancer as well as tools in regenerative medicine and fundamental science. In recent years, there have been several reports claiming the identification of small molecule agents that are selective toward certain FZD proteins using a variety of approaches. However, the majority of these studies lacked a selective functional assay to validate their functionality. In this study, we describe the development and application of a selective assay for individual FZD proteins. Our findings indicate that the majority of reported compounds lack the capacity to inhibit the functioning of the claimed FZD proteins when stimulated by a Wnt ligand in the canonical pathway. Instead, the compounds demonstrate a broad range of off-target effects, including inhibition of downstream pathway component(s) (3235-0367, SRI35959, carbamazepine, niclosamide), lack of activity (FzM1), and surprising antagonism of firefly luciferase (F7H). The only compound that fulfills the expected selectivity profile is peptide Fz7-21. These results highlight the necessity of implementing rigorous testing of the screening-derived compounds in selective functional assays and are important for the field of drug discovery and development targeting the highly demanded Wnt-FZD pathway.
Collapse
Affiliation(s)
- Alexey Koval
- Department of Cell Physiology
and Metabolism, Translational Research Centre in Oncohaematology,
Faculty of Medicine, University of Geneva,1206 Geneva, Switzerland
| | - Cédric Boudou
- Department of Cell Physiology
and Metabolism, Translational Research Centre in Oncohaematology,
Faculty of Medicine, University of Geneva,1206 Geneva, Switzerland
| | - Vladimir L. Katanaev
- Department of Cell Physiology
and Metabolism, Translational Research Centre in Oncohaematology,
Faculty of Medicine, University of Geneva,1206 Geneva, Switzerland
| |
Collapse
|
39
|
Ntourmas S, Sachs M, Paclíková P, Brückner M, Bryja V, Behrens J, Bernkopf DB. Endogenous oligomer formation underlies DVL2 condensates and promotes Wnt/β-catenin signaling. eLife 2024; 13:RP96841. [PMID: 39652469 PMCID: PMC11627551 DOI: 10.7554/elife.96841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
Activation of the Wnt/β-catenin pathway crucially depends on the polymerization of dishevelled 2 (DVL2) into biomolecular condensates. However, given the low affinity of known DVL2 self-interaction sites and its low cellular concentration, it is unclear how polymers can form. Here, we detect oligomeric DVL2 complexes at endogenous protein levels in human cell lines, using a biochemical ultracentrifugation assay. We identify a low-complexity region (LCR4) in the C-terminus whose deletion and fusion decreased and increased the complexes, respectively. Notably, LCR4-induced complexes correlated with the formation of microscopically visible multimeric condensates. Adjacent to LCR4, we mapped a conserved domain (CD2) promoting condensates only. Molecularly, LCR4 and CD2 mediated DVL2 self-interaction via aggregating residues and phenylalanine stickers, respectively. Point mutations inactivating these interaction sites impaired Wnt pathway activation by DVL2. Our study discovers DVL2 complexes with functional importance for Wnt/β-catenin signaling. Moreover, we provide evidence that DVL2 condensates form in two steps by pre-oligomerization via high-affinity interaction sites, such as LCR4, and subsequent condensation via low-affinity interaction sites, such as CD2.
Collapse
Affiliation(s)
- Senem Ntourmas
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-NürnbergErlangenGermany
| | - Martin Sachs
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-NürnbergErlangenGermany
| | - Petra Paclíková
- Department of Experimental Biology, Faculty of Science, Masaryk UniversityBrnoCzech Republic
| | - Martina Brückner
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-NürnbergErlangenGermany
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk UniversityBrnoCzech Republic
| | - Jürgen Behrens
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-NürnbergErlangenGermany
| | - Dominic B Bernkopf
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-NürnbergErlangenGermany
| |
Collapse
|
40
|
Xiao D, Liu S, Xiang M. Unveiling the potential: implications of successful somatic cell-to-ganglion organoid reprogramming. Curr Opin Genet Dev 2024; 89:102227. [PMID: 39586653 DOI: 10.1016/j.gde.2024.102227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/30/2024] [Accepted: 06/24/2024] [Indexed: 11/27/2024]
Abstract
Organoids have a wide range of potential applications in areas such as organ development, precision medicine, regenerative medicine, drug screening, disease modeling, and gene editing. Currently, most organoids are generated through three-dimensional (3D) in vitro culture of adult stem cells or pluripotent stem cells. However, this method of generating organoids still has several limitations and challenges, including complex manipulations, costly culturing materials, extended time requirements, and certain heterogeneity. Recently, we have found that fibroblasts, when overexpressing several key regulatory transcription factors, are able to directly and rapidly generate two types of ganglion organoids: sensory ganglion (SG) and autonomic ganglion (AG) organoids. They have structures and electrophysiological properties similar to those of endogenous organs in the body. Here, we provide a brief overview of organoid development, focusing on direct reprogramming of SG and AG organoids and their transplantation and regeneration. Finally, the advantages and prospects of direct reprogramming of organoids are discussed.
Collapse
Affiliation(s)
- Dongchang Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Shuting Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Mengqing Xiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sxen University, Guangzhou 510080, China.
| |
Collapse
|
41
|
Tamtaji OR, Ostadian A, Homayoonfal M, Nejati M, Mahjoubin-Tehran M, Nabavizadeh F, Ghelichi E, Mohammadzadeh B, Karimi M, Rahimian N, Mirzaei H. Cerium(IV) oxide:silver/graphene oxide (CeO2:Ag/GO) nanoparticles modulate gene expression and inhibit colorectal cancer cell growth: a pathway-centric therapeutic approach. Cancer Nanotechnol 2024; 15:62. [DOI: 10.1186/s12645-024-00300-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/18/2024] [Indexed: 01/06/2025] Open
|
42
|
Zhang S, Guo Y, Lu Y, Liu F, Heng BC, Deng X. The considerations on selecting the appropriate decellularized ECM for specific regeneration demands. Mater Today Bio 2024; 29:101301. [PMID: 39498148 PMCID: PMC11532911 DOI: 10.1016/j.mtbio.2024.101301] [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: 08/10/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
An ideal biomaterial should create a customized tissue-specific microenvironment that can facilitate and guide the tissue repair process. Due to its good biocompatibility and similar biochemical properties to native tissues, decellularized extracellular matrix (dECM) generally yields enhanced regenerative outcomes, with improved morphological and functional recovery. By utilizing various decellularization techniques and post-processing protocols, dECM can be flexibly prepared in different states from various sources, with specifically customized physicochemical properties for different tissues. To initiate a well-orchestrated tissue-regenerative response, dECM exerts multiple effects at the wound site by activating various overlapping signaling pathways to promote cell adhesion, proliferation, and differentiation, as well as suppressing inflammation via modulation of various immune cells, including macrophages, T cells, and mastocytes. Functional tissue repair is likely the main aim when employing the optimized dECM biomaterials. Here, we review the current applications of different kinds of dECMs in an attempt to improve the efficiency of tissue regeneration, highlighting key considerations on developing dECM for specific tissue engineering applications.
Collapse
Affiliation(s)
- Shihan Zhang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Yaru Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Yixuan Lu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Fangyong Liu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- NMPA Key Laboratory for Dental Materials, Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| |
Collapse
|
43
|
Li Y, Wang X, Huang M, Wang X, Li C, Li S, Tang Y, Yu S, Wang Y, Song W, Wu W, Liu Y, Chen YG. BMP suppresses Wnt signaling via the Bcl11b-regulated NuRD complex to maintain intestinal stem cells. EMBO J 2024; 43:6032-6051. [PMID: 39433900 PMCID: PMC11612440 DOI: 10.1038/s44318-024-00276-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 09/22/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024] Open
Abstract
Lgr5+ intestinal stem cells (ISCs) are crucial for the intestinal epithelium renewal and regeneration after injury. However, the mechanism underlying the interplay between Wnt and BMP signaling in this process is not fully understood. Here we report that Bcl11b, which is downregulated by BMP signaling, enhances Wnt signaling to maintain Lgr5+ ISCs and thus promotes the regeneration of the intestinal epithelium upon injury. Loss of Bcl11b function leads to a significant decrease of Lgr5+ ISCs in both intestinal crypts and cultured organoids. Mechanistically, BMP suppresses the expression of Bcl11b, which can positively regulate Wnt target genes by inhibiting the function of the Nucleosome Remodeling and Deacetylase (NuRD) complex and facilitating the β-catenin-TCF4 interaction. Bcl11b can also promote intestinal epithelium repair after injuries elicited by both irradiation and DSS-induced inflammation. Furthermore, Bcl11b deletion prevents proliferation and tumorigenesis of colorectal cancer cells. Together, our findings suggest that BMP suppresses Wnt signaling via Bcl11b regulation, thus balancing homeostasis and regeneration in the intestinal epithelium.
Collapse
Affiliation(s)
- Yehua Li
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiaodan Wang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Meimei Huang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xu Wang
- Guangzhou National Laboratory, Guangzhou, 510700, China
| | - Chunlin Li
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Siqi Li
- Guangzhou National Laboratory, Guangzhou, 510700, China
| | - Yuhui Tang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Shicheng Yu
- Guangzhou National Laboratory, Guangzhou, 510700, China
| | - Yalong Wang
- Guangzhou National Laboratory, Guangzhou, 510700, China
| | - Wanglu Song
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Wei Wu
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuan Liu
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Guangzhou National Laboratory, Guangzhou, 510700, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
44
|
Qin T, Han J, Fan C, Sun H, Rauf N, Wang T, Yin Z, Chen X. Unveiling axolotl transcriptome for tissue regeneration with high-resolution annotation via long-read sequencing. Comput Struct Biotechnol J 2024; 23:3186-3198. [PMID: 39263210 PMCID: PMC11388199 DOI: 10.1016/j.csbj.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/13/2024] Open
Abstract
Axolotls are known for their remarkable regeneration ability. Exploring their transcriptome provides insight into regenerative mechanisms. However, the current annotation of the axolotl transcriptome is limited, leaving the role of unannotated transcripts in regeneration unknown. To discourse this challenge, we exploited long-read sequencing technology, which enables direct observation of full-length RNA transcripts, greatly enhancing the coverage and accuracy of axolotl transcriptome annotation. By utilizing this method, we identified 222 novel gene loci and 4775 novel transcripts, which were quantified using short-read sequencing data. Through the inclusive analysis, we discovered novel homologs, potential functional proteins, noncoding RNAs, and alternative splicing events in key regeneration pathways. In particular, we identified novel transcripts with high protein-coding potential implicated in cell cycle regulation and musculoskeletal development, and regeneration were identified. Interestingly, alternative splice variants were also detected across diverse pathways critical to regeneration. This specifies that these novel transcripts potentially play vital roles underpinning the robust regenerative capacities of axolotls. Single-cell transcriptomic analysis further revealed these isoforms to predominantly exist in axolotl limb chondrocytes and mature tissue cell populations. Overall, the findings significantly advanced consideration of the axolotl transcriptome and provided a new perspective for understanding the mechanisms of regenerative abilities of axolotls.
Collapse
Affiliation(s)
- Tian Qin
- Department of Orthopedic Surgery of Sir Run Run Shaw Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Jie Han
- Department of Sports Medicine & Orthopedic Surgery, The Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunmei Fan
- Department of Sports Medicine & Orthopedic Surgery, The Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Heng Sun
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, China
| | - Naveed Rauf
- Department of Sports Medicine & Orthopedic Surgery, The Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Tingzhang Wang
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Zi Yin
- Department of Orthopedic Surgery of Sir Run Run Shaw Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Xiao Chen
- Department of Sports Medicine & Orthopedic Surgery, The Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| |
Collapse
|
45
|
Fisch AS, Pestana A, Sachse V, Doll C, Hofmann E, Heiland M, Obermueller T, Heidemann J, Dommerich S, Schoppe D, Schallenberg S, Piwonski I, Blanc E, Tinhofer I. Feasibility analysis of using patient-derived tumour organoids for treatment decision guidance in locally advanced head and neck squamous cell carcinoma. Eur J Cancer 2024; 213:115100. [PMID: 39476443 DOI: 10.1016/j.ejca.2024.115100] [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: 09/21/2024] [Accepted: 10/20/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Current treatment for head and neck squamous cell carcinoma (HNSCC) involves surgery, radiotherapy, and chemotherapy. Despite aggressive multimodal approaches, tumour recurrence occurs in 40-60 % of cases, leading to poor survival outcomes. HNSCC lacks common genetic drivers for tailored therapies, and reliable biomarkers for treatment selection are scarce. We investigated the procedural requirements for incorporating drug- and radiosensitivity screens in patient-derived organoids (PDOs) within a clinical trial framework. PATIENTS AND METHODS Fresh tumour samples (N = 198) from 186 HNSCC patients were included. Success rates of organoid establishment were correlated with clinical and procedural parameters. Timelines for establishment of PDO cultures were determined, and their long-term growth potential assessed by serial passaging. Additionally, we conducted whole exome sequencing on matched tumour-organoid pairs. Three PDO models were employed to establish radiosensitivity assays. RESULTS In total, PDO models displaying histomorphological features and genomic alterations of parental tumours were successfully established for 35 % of patient tumours. Success rates rose to 77 % for samples with a tumour cell content of 30 % or higher. Advanced patient age, prior radiotherapy, and delays in tissue processing were identified as negative predictors for engraftment. The estimated time interval needed for screens was compatible with PDO-guided selection of curative-intent radiotherapy regimens. CONCLUSIONS Our findings suggest that with high-quality samples and efficient tissue processing, PDO screens can be successfully performed in 77 % of HNSCC patients. Given the procedural challenges involved, future clinical trials aiming to the utility of PDOs for guiding treatment decisions should consider implementing centralised PDO screening.
Collapse
Affiliation(s)
- Anne-Sophie Fisch
- Department of Radiooncology and Radiotherapy, Translational Radiation Oncology Research Laboratory, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Ana Pestana
- Department of Radiooncology and Radiotherapy, Translational Radiation Oncology Research Laboratory, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; German Cancer Research Center (DKFZ), Heidelberg, and German Cancer Consortium (DKTK), partner site Berlin, Germany
| | - Vanessa Sachse
- Department of Radiooncology and Radiotherapy, Translational Radiation Oncology Research Laboratory, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; German Cancer Research Center (DKFZ), Heidelberg, and German Cancer Consortium (DKTK), partner site Berlin, Germany
| | - Christian Doll
- Department of Oral and Maxillofacial Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Elena Hofmann
- Department of Oral and Maxillofacial Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Max Heiland
- Department of Oral and Maxillofacial Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Theresa Obermueller
- Department of Otorhinolaryngology, Campus Benjamin-Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Jan Heidemann
- Department of Otorhinolaryngology, Campus Benjamin-Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Steffen Dommerich
- Department of Otorhinolaryngology, Campus Mitte, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Diana Schoppe
- Department of Radiooncology and Radiotherapy, Translational Radiation Oncology Research Laboratory, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Simon Schallenberg
- Institut für Pathologie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Iris Piwonski
- Institut für Pathologie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Eric Blanc
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Ingeborg Tinhofer
- Department of Radiooncology and Radiotherapy, Translational Radiation Oncology Research Laboratory, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; German Cancer Research Center (DKFZ), Heidelberg, and German Cancer Consortium (DKTK), partner site Berlin, Germany.
| |
Collapse
|
46
|
He R, Wei Y, Yan S, Chen J, Guan Y, Xiong X, Liang L, Guan C, Liu H, Ouyang Y, Wang J, Peng X, Ye J, Zhao J, Lai B, Wang Y, Peng J, Quan Q. Wnt 3a-Modified Scaffolds Improve Nerve Regeneration by Boosting Schwann Cell Function. ACS APPLIED MATERIALS & INTERFACES 2024; 16:63317-63332. [PMID: 39520323 PMCID: PMC11583969 DOI: 10.1021/acsami.4c15013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
A pivotal approach in engineering artificial peripheral nerve sheaths encompasses the augmentation of the regenerative microenvironment via the manipulation of Schwann cells (SCs). Our investigation employed single-cell sequencing analysis to elucidate the potential functions of Schwann cells and the Wnt pathway in facilitating peripheral nerve regeneration. In vitro studies showed that activating the Wnt signaling pathway promotes the transition to repair SCs, boosting their growth, movement, and immune functions. To better understand the peripheral nerve regeneration environment, we created a polymer scaffold using ammonization and electrospinning. The Wnt3a protein was incorporated into the polycaprolactone (PCL) electrospun fiber surface. In a rat sciatic nerve defect model, the Wnt3a-modified scaffold showed better nerve repair outcomes than traditional electrospun scaffolds. After a week, the test group showed better immune regulation and angiogenesis, with a significant increase in axon growth rate observed after 3 weeks. Three-month-long animal experiments revealed notable improvements in neuroelectrophysiology, reduced organ atrophy, and enhanced sciatic nerve recovery. In this nerve defect model, Wnt3a-modified neural scaffolds achieved repair effects.
Collapse
Affiliation(s)
- Ruichao He
- School of Medicine, Nankai University, Tianjin 300071, P. R. China
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Yu Wei
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Shi Yan
- School of Medicine, Nankai University, Tianjin 300071, P. R. China
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Jiajie Chen
- School of Medicine, Nankai University, Tianjin 300071, P. R. China
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Yanjun Guan
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Xing Xiong
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Lijing Liang
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Congcong Guan
- School of Medicine, Nankai University, Tianjin 300071, P. R. China
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Haolin Liu
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Yiben Ouyang
- School of Medicine, Nankai University, Tianjin 300071, P. R. China
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Junli Wang
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Xiwei Peng
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Jianting Ye
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Jinjuan Zhao
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| | - Biqin Lai
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou 510080, P. R. China
| | - Yu Wang
- School of Medicine, Nankai University, Tianjin 300071, P. R. China
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province 226007, P. R. China
| | - Jiang Peng
- School of Medicine, Nankai University, Tianjin 300071, P. R. China
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province 226007, P. R. China
| | - Qi Quan
- Department of Orthopedic Surgery, Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing,100048, P. R. China
| |
Collapse
|
47
|
Cheng X, Shen H, Zhang W, Chen B, Xu S, Wu L. Characterizing the effects of triclosan and triclocarban on the intestinal epithelial homeostasis using small intestinal organoids. JOURNAL OF HAZARDOUS MATERIALS 2024; 479:135734. [PMID: 39244982 DOI: 10.1016/j.jhazmat.2024.135734] [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: 06/06/2024] [Revised: 08/02/2024] [Accepted: 09/01/2024] [Indexed: 09/10/2024]
Abstract
Intestinal epithelium has the largest surface of human body, contributes dramatically to defense of toxicant-associated intestinal injury. Triclosan (TCS) and triclocarban (TCC), extensively employed as antibacterial agents in personal care products (PCPs) and healthcare facilities, caused serious damage to human intestine. However, the role of the intestinal epithelium in TCS/TCC-induced intestinal toxicity and its underlying toxic mechanisms remain incompletely understood. In this study, a novel 3D intestinal organoid model was utilized to investigate that exposure to TCS/TCC led to a compromised self-renewal and differentiation of intestinal stem cells (ISCs). Consequently, this disrupted intestinal epithelial homeostasis ultimately caused a reduction in nutrient absorption and deficient of epithelial defense to exogenous and endogenous pathogens stimulation. The inhibition of the Wnt signaling pathway in intestinal stem cell was contributed to the intestinal toxicity of TCS/TCC. These results were further confirmed in vivo with mice exposed to TCS/TCC. The findings of this study provide evidence that TCS/TCC possess the capacity to disturb the homeostasis of the intestinal epithelium, and emphasize the credibility of organoids as a valuable model for toxicological studies, as they could faithfully recapitulate in vivo phenomena.
Collapse
Affiliation(s)
- Xiaowen Cheng
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, PR China
| | - Hongzhi Shen
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, PR China
| | - Wen Zhang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, PR China
| | - Biao Chen
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, PR China
| | - Shengmin Xu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, PR China.
| | - Lijun Wu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, PR China
| |
Collapse
|
48
|
Jin B, Su G, Zhou X, Xu L, Wang W, Zhou T, Tan Y, Wang S, Li G. Basic Fibroblast Growth Factor Supports the Function of Limbal Niche Cells via the Wnt/β-Catenin Pathway. J Ocul Pharmacol Ther 2024; 40:571-580. [PMID: 39083404 DOI: 10.1089/jop.2024.0042] [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] [Indexed: 08/02/2024] Open
Abstract
Purpose: To test the effects and underlying mechanisms of basic fibroblast growth factor (bFGF) on the limbal niche cell (LNC) function ex vivo. Methods: By using different concentrations of bFGF (0, 4, 8, 12, and 16 ng/mL) and fibroblast growth factor receptor (FGFR) inhibitors, the effects of bFGF on LNC proliferation, expression of stem cell markers, and transcription levels of the β-catenin were investigated. Single-cell RNA sequencing (scRNA-seq) was used to analyze the action and mechanisms of FGFR subtypes and the Wnt/β-catenin pathway during LNC culture. An mature corneal epithelial cell (MCEC)/LNC three-dimensional model was constructed to verify whether bFGF activates the Wnt/β-catenin pathway in LNC by inhibiting FGFR or β-catenin targets. Results: scRNA-seq showed that FGFR1 is the main receptor in LNC, along with the molecules in the Wnt pathway, including WNT2, FZD7, LRP5, LRP6, and β-catenin. The 12 ng/mL bFGF treatment group showed higher LNC proliferation rate and transcription levels of OCT4, SOX2, NANOG, and β-catenin than any other groups (P < 0.001). In the MCEC/LNC co-culture model, MCEC/LNC treated with 12 ng/mL bFGF promoted the aggregation of the spheres than other groups, associated with increased transcription levels of P63α, WNT2, β-catenin, and a decreased transcription level of CK12 (P < 0.001). Wnt/β-catenin inhibitor LF3 treatment reversed the abovementioned effect of bFGF. Conclusions: bFGF could maintain and promote the stemness of LNC via the FGFR1/Wnt2/FZD7/LRP6 axis in a concentration-dependent manner.
Collapse
Affiliation(s)
- Bihui Jin
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanyu Su
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Zhou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingjuan Xu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Zhou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongyao Tan
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shusheng Wang
- Department of Cell and Molecular Biology & Ophthalmology, Tulane University, New Orleans, Louisiana, USA
| | - Guigang Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
49
|
Wang X, Zou K, Xiong Y, Zheng Y, Zheng J, Liu Y, Zhong T, Zhao X. Dietary titanium dioxide nanoparticles impair intestinal epithelial regeneration by perturbating the function of intestinal stem cells. Food Chem Toxicol 2024; 193:115057. [PMID: 39406333 DOI: 10.1016/j.fct.2024.115057] [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: 05/12/2024] [Revised: 07/01/2024] [Accepted: 10/12/2024] [Indexed: 10/19/2024]
Abstract
Intestinal health is closely linked to intestinal stem cells (ISCs), which are highly sensitive to the harmful substances in the lumen. However, there is limited knowledge regarding the effects of food additives on ISCs. This study aims to investigate the impact of dietary titanium dioxide nanoparticles (TiO2 NPs) compared with titanium dioxide microparticles (TiO2 MPs) on intestinal health associated with ISCs in response to dextran sodium sulfate (DSS)-induced enteritis in mice, as well as the related mechanism. We found that exposure to 1% (w/w) TiO2 NPs aggravated DSS-induced enteritis in mice, while this effect could not be observed under exposure to TiO2 MPs. Additionally, 1% (w/w) TiO2 NPs exposure under DSS-induced enteritis worsened the ISC-mediated regeneration of intestinal epithelium by decreasing the epithelial cell proliferation and epithelial turnover rate while increasing epithelial cell death. Meanwhile, using a 3D intestinal organoid model, we discovered that 20 μg/mL TiO2 NPs impaired ISC function and disrupted ISC fate specification both ex vivo and in vitro. Furthermore, TiO2 NPs hindered the nuclear translocation of β-catenin, reducing the overall output of Wnt signaling. Together, TiO2 NPs deteriorated the intestinal epithelial regeneration of mice with DSS-induced enteritis by perturbating ISC function and fate specification through a mechanism involving Wnt signaling. These findings highlight the adverse effect of dietary TiO2 NPs on ISCs and shed light on the particle size optimization of TiO2 food additive.
Collapse
Affiliation(s)
- Xiu Wang
- School of Advanced Materials Engineering, Jiaxing Nanhu University, Jiaxing, 314000, China; Jiaxing Key Laboratory for Research and Application of Green and Low-carbon Advanced Materials, Jiaxing, 314000, China.
| | - Kai Zou
- School of Advanced Materials Engineering, Jiaxing Nanhu University, Jiaxing, 314000, China; Jiaxing Key Laboratory for Research and Application of Green and Low-carbon Advanced Materials, Jiaxing, 314000, China
| | - Yu Xiong
- School of Advanced Materials Engineering, Jiaxing Nanhu University, Jiaxing, 314000, China
| | - Yongwang Zheng
- School of Advanced Materials Engineering, Jiaxing Nanhu University, Jiaxing, 314000, China
| | - Jiale Zheng
- School of Advanced Materials Engineering, Jiaxing Nanhu University, Jiaxing, 314000, China
| | - Yong Liu
- School of Advanced Materials Engineering, Jiaxing Nanhu University, Jiaxing, 314000, China
| | - Ting Zhong
- School of Advanced Materials Engineering, Jiaxing Nanhu University, Jiaxing, 314000, China
| | - Xincheng Zhao
- School of Advanced Materials Engineering, Jiaxing Nanhu University, Jiaxing, 314000, China
| |
Collapse
|
50
|
Chen S, Liu Y, Chen X, Tao H, Piao Y, Huang H, Han Z, Han Z, Chen X, Li Z. Combined lineage tracing and scRNA-seq reveal the activation of Sox9 + cells in renal regeneration with PGE 2 treatment. Cell Prolif 2024; 57:e13679. [PMID: 38801100 PMCID: PMC11533080 DOI: 10.1111/cpr.13679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/07/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
Uncovering mechanisms of endogenous regeneration and repair through resident stem cell activation will allow us to develop specific therapies for injuries and diseases by targeting resident stem cell lineages. Sox9+ stem cells have been reported to play an essential role in acute kidney injury (AKI). However, a complete view of the Sox9+ lineage was not well investigated to accurately elucidate the functional end state and the choice of cell fate during tissue repair after AKI. To identify the mechanisms of fate determination of Sox9+ stem cells, we set up an AKI model with prostaglandin E2 (PGE2) treatment in a Sox9 lineage tracing mouse model. Single-cell RNA sequencing (scRNA-seq) was performed to analyse the transcriptomic profile of the Sox9+ lineage. Our results revealed that PGE2 could activate renal Sox9+ cells and promote the differentiation of Sox9+ cells into renal proximal tubular epithelial cells and inhibit the development of fibrosis. Furthermore, single-cell transcriptome analysis demonstrated that PGE2 could regulate the restoration of lipid metabolism homeostasis in proximal tubular epithelial cells by participating in communication with different cell types. Our results highlight the prospects for the activation of endogenous renal Sox9+ stem cells with PGE2 for the regenerative therapy of AKI.
Collapse
Affiliation(s)
- Shang Chen
- Nankai University School of MedicineTianjinChina
- The Key Laboratory of Bioactive Materials, Ministry of EducationNankai UniversityTianjinChina
| | - Yue Liu
- Nankai University School of MedicineTianjinChina
| | - Xiaoniao Chen
- Department of OphthalmologyThe Third Medical Center of Chinese PLA General HospitalBeijingChina
- National Key Laboratory of Kidney DiseasesChinese PLA General HospitalBeijingChina
| | - Hongyan Tao
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Yongjun Piao
- Nankai University School of MedicineTianjinChina
| | - Haoyan Huang
- Nankai University School of MedicineTianjinChina
| | - Zhibo Han
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjinChina
- Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center for Cell ProductsAmCellGene Co., Ltd.TianjinChina
| | - Zhong‐Chao Han
- Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center for Cell ProductsAmCellGene Co., Ltd.TianjinChina
| | - Xiang‐Mei Chen
- National Key Laboratory of Kidney DiseasesChinese PLA General HospitalBeijingChina
| | - Zongjin Li
- Nankai University School of MedicineTianjinChina
- The Key Laboratory of Bioactive Materials, Ministry of EducationNankai UniversityTianjinChina
- National Key Laboratory of Kidney DiseasesChinese PLA General HospitalBeijingChina
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology ObstetricsNankai University Affiliated Hospital of Obstetrics and GynecologyTianjinChina
- Henan Key Laboratory of Cardiac Remodeling and TransplantationZhengzhou No. 7 People's HospitalZhengzhouChina
| |
Collapse
|