1
|
Plessis-Belair J, Russo T, Riessland M, Sher RB. Nuclear Import Defects Drive Cell Cycle Dysregulation in Neurodegeneration. Aging Cell 2025:e70091. [PMID: 40377023 DOI: 10.1111/acel.70091] [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: 01/24/2025] [Revised: 03/31/2025] [Accepted: 04/14/2025] [Indexed: 05/18/2025] Open
Abstract
Neurodegenerative diseases (NDDs) and other age-related disorders have been classically defined by a set of key pathological hallmarks. Two of these hallmarks, cell cycle dysregulation (CCD) and nucleocytoplasmic transport (NCT) defects, have long been debated as being either causal or consequential in the pathology of accelerated aging. Specifically, aberrant cell cycle activation in post-mitotic neurons has been shown to trigger neuronal cell death pathways and cellular senescence. Additionally, NCT has been observed to be progressively dysregulated during aging and in neurodegeneration, where the increased subcellular redistribution of nuclear proteins, such as TAR DNA-Binding Protein-43 (TDP-43), to the cytoplasm is a primary driver of disease. However, the functional significance of NCT defects as either a causal mechanism or consequence of pathology, and how the redistribution of cell cycle machinery contributes to neurodegeneration, remains unclear. Here, we describe that pharmacological inhibition of importin-β nuclear import is capable of perturbing cell cycle machinery both in mitotic neuronal cell lines and post-mitotic primary neurons in vitro. Our NemfR86S mouse model of motor neuron disease, characterized by nuclear import defects, further recapitulates the hallmarks of CCD we observed in mitotic cell lines and in post-mitotic primary neurons in vitro, and in spinal motor neurons in vivo. The observed CCD is consistent with the transcriptional and phenotypical dysregulation commonly associated with neuronal cell death and senescence-like features in NDDs. Together, this evidence suggests that impairment of nuclear import pathways resulting in CCD may be a common driver of pathology in neurodegeneration.
Collapse
Affiliation(s)
- Jonathan Plessis-Belair
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, New York, USA
| | - Taylor Russo
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, New York, USA
| | - Markus Riessland
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, New York, USA
| | - Roger B Sher
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
2
|
Militi S, Nibhani R, Pook M, Pauklin S. SMAD2/3-SMYD2 and developmental transcription factors cooperate with cell-cycle inhibitors to guide tissue formation. Protein Cell 2025; 16:260-285. [PMID: 38758030 PMCID: PMC12053477 DOI: 10.1093/procel/pwae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/05/2024] [Indexed: 05/18/2024] Open
Abstract
Tissue formation and organ homeostasis are achieved by precise coordination of proliferation and differentiation of stem cells and progenitors. While deregulation of these processes can result in degenerative disease or cancer, their molecular interplays remain unclear. Here we show that the switch of human pluripotent stem cell (hPSC) self-renewal to differentiation is associated with the induction of distinct cyclin-dependent kinase inhibitors (CDKIs). In hPSCs, Activin/Nodal/TGFβ signaling maintains CDKIs in a poised state via SMAD2/3-NANOG-OCT4-EZH2-SNON transcriptional complex. Upon gradual differentiation, CDKIs are induced by successive transcriptional complexes between SMAD2/3-SMYD2 and developmental regulators such as EOMES, thereby lengthening the G1 phase. This, in turn, induces SMAD2/3 transcriptional activity by blocking its linker phosphorylation. Such SMAD2/3-CDKI positive feedback loops drive the exit from pluripotency and stepwise cell-fate specification that could be harnessed for producing cells for therapeutic applications. Our study uncovers fundamental mechanisms of how cell-fate specification is interconnected to cell-cycle dynamics and provides insight into autonomous circuitries governing tissue self-formation.
Collapse
Affiliation(s)
- Stefania Militi
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road, Headington, Oxford OX3 7LD, United Kingdom
| | - Reshma Nibhani
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road, Headington, Oxford OX3 7LD, United Kingdom
| | - Martin Pook
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road, Headington, Oxford OX3 7LD, United Kingdom
| | - Siim Pauklin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road, Headington, Oxford OX3 7LD, United Kingdom
| |
Collapse
|
3
|
Jahanshir E, Llamas J, Kim Y, Biju K, Oak S, Gnedeva K. The Hippo pathway and p27 Kip1 cooperate to suppress mitotic regeneration in the organ of Corti and the retina. Proc Natl Acad Sci U S A 2025; 122:e2411313122. [PMID: 40178894 PMCID: PMC12002246 DOI: 10.1073/pnas.2411313122] [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: 06/17/2024] [Accepted: 02/19/2025] [Indexed: 04/05/2025] Open
Abstract
The mature mammalian auditory sensory organ, the organ of Corti (OC), lacks the capacity for regenerating hair cells, leading to permanent hearing impairment. In contrast, the vestibular system has a limited capacity for hair cell regeneration, which we have shown to be further enhanced by inhibiting the Hippo pathway. Here, we demonstrate that, despite similar transcriptional responses, only vestibular and not auditory supporting cells proliferate as a result of Yap activation following Hippo inhibition. Mechanistically, we identify p27Kip1, a cell cycle kinase inhibitor encoded by Cdkn1b, as an additional barrier preventing cell cycle reentry specifically in the OC. We show that while in both systems Yap stimulates p27Kip1 degradation through activation of its direct target gene Skp2, this protein-level control is antagonized by an unusually high level of Cdkn1b transcription in the cochlea. Consequently, p27Kip1 activity is maintained in the OC even in the presence of constitutively active Yap5SA, counteracting its mitogenic effects. Supporting this model, inactivation of the Hippo pathway in the Cdkn1b-deficient background is sufficient to induce adult auditory supporting cell proliferation in vivo. Furthermore, we show that the synergistic interaction between Hippo and p27Kip1 is conserved in the retina where inhibition of both pathways potently induces Müller glia proliferation and initiates neuronal regeneration. Our work uncovers the molecular mechanism preventing quiescent adult sensory progenitor cells, supporting cells in the ear and Müller glia in the eye, from reentering the cell cycle after damage-the key step toward sensory receptor regeneration blocked in mammals.
Collapse
Affiliation(s)
- Eva Jahanshir
- University of Southern California Caruso Department of Otolaryngology—Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Juan Llamas
- University of Southern California Caruso Department of Otolaryngology—Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Yeeun Kim
- University of Southern California Caruso Department of Otolaryngology—Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Kevin Biju
- University of Southern California Caruso Department of Otolaryngology—Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Sanyukta Oak
- University of Southern California Caruso Department of Otolaryngology—Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Ksenia Gnedeva
- University of Southern California Caruso Department of Otolaryngology—Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| |
Collapse
|
4
|
Schirripa A, Schöppe H, Nebenfuehr S, Zojer M, Klampfl T, Kugler V, Maw BS, Ceylan H, Uras IZ, Scheiblecker L, Gamper E, Stelzl U, Stefan E, Kaserer T, Sexl V, Kollmann K. Cdk6's functions are critically regulated by its unique C-terminus. iScience 2025; 28:111697. [PMID: 39898030 PMCID: PMC11787673 DOI: 10.1016/j.isci.2024.111697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/09/2024] [Accepted: 12/24/2024] [Indexed: 02/04/2025] Open
Abstract
The vital cell cycle machinery is tightly regulated and alterations of its central signaling hubs are a hallmark of cancer. The activity of CDK6 is controlled by interaction with several partners including cyclins and INK4 proteins, which have been shown to mainly bind to the amino-terminal lobe. We analyzed the impact of CDK6's C-terminus on its functions in a leukemia model, revealing a central role in promoting proliferation. C-terminally truncated Cdk6 (Cdk6 ΔC) shows reduced nuclear translocation and therefore chromatin interaction and fails to enhance proliferation and disease progression. The combination of proteomic analysis and protein modeling highlights that Cdk6's C-terminus is essential for protein flexibility and for its binding potential to cyclin D, p27Kip1 and INK4 proteins but not cyclin B. We demonstrate that the C-terminus is a unique and essential part of the CDK6 protein, regulating interaction partner binding and therefore CDK6's functionality.
Collapse
Affiliation(s)
- Alessia Schirripa
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Helge Schöppe
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Sofie Nebenfuehr
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Markus Zojer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Thorsten Klampfl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Valentina Kugler
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020 Innsbruck, Austria
- Institute of Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Belinda S. Maw
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Huriye Ceylan
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Iris Z. Uras
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Lisa Scheiblecker
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Elisabeth Gamper
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Ulrich Stelzl
- Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Field of Excellence BioHealth - University of Graz, Graz, Austria
| | - Eduard Stefan
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020 Innsbruck, Austria
- Institute of Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Teresa Kaserer
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
- University of Innsbruck, Innsbruck, Austria
| | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| |
Collapse
|
5
|
Plessis-Belair J, Russo T, Riessland M, Sher RB. Nuclear Import Defects Drive Cell Cycle Dysregulation in Neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635269. [PMID: 39975276 PMCID: PMC11838365 DOI: 10.1101/2025.01.28.635269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Neurodegenerative diseases (NDDs) and other age-related disorders have been classically defined by a set of key pathological hallmarks. Two of these hallmarks, cell cycle dysregulation (CCD) and nucleocytoplasmic transport (NCT) defects, have long been debated as being either causal or consequential in the pathology of accelerated aging. Specifically, aberrant cell cycle activation in post-mitotic neurons has been shown to trigger neuronal cell death pathways and cellular senescence. Additionally, NCT has been observed to be progressively dysregulated during aging and in neurodegeneration, where the increased subcellular redistribution of nuclear proteins such as TAR DNA-Binding Protein-43 (TDP43) to the cytoplasm is a primary driver of many NDDs. However, the functional significance of NCT defects as either a primary driver or consequence of pathology, and how the redistribution of cell cycle machinery contributes to neurodegeneration, remains unclear. Here, we describe that pharmacological inhibition of importin-β nuclear import is capable of perturbing cell cycle machinery both in mitotic neuronal cell lines and post-mitotic primary neurons in vitro. Our Nemf R86S mouse model of motor neuron disease, characterized by nuclear import defects, further recapitulates the hallmarks of CCD in mitotic cell lines and in post-mitotic primary neurons in vitro, and in spinal motor neurons in vivo. The observed CCD is consistent with the transcriptional and phenotypical dysregulation observed in neuronal cell death and cellular senescence in NDDs. Together, this evidence suggests that impairment of nuclear import pathways resulting in CCD may be a common driver of pathology in neurodegeneration.
Collapse
Affiliation(s)
- Jonathan Plessis-Belair
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| | - Taylor Russo
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| | - Markus Riessland
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| | - Roger B Sher
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
6
|
Zou J, Wang D, Yin G, Lu K, Chang K, Li H. Prognostic significance of p27 in colorectal cancer: a meta-analysis and bioinformatics analysis. Front Oncol 2024; 14:1495476. [PMID: 39845325 PMCID: PMC11751620 DOI: 10.3389/fonc.2024.1495476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/29/2024] [Indexed: 01/24/2025] Open
Abstract
Background In the past, numerous investigations have delved into the influence of p27 (p27kip) on the prognosis and clinicopathological characteristics of colorectal cancer (CRC), yielding conclusions that are not universally statistically significant, thus rendering the discourse rather contentious. Methods We collected available articles published before August 2024 and extracted data to analyze the association between the expression of p27 and the prognosis and clinicopathological features of CRC. In addition, we used Gene Expression Profiling Interactive Analysis (GEPIA), University of Alabama at Birmingham's Cancer Data Analysis Portal (UALCAN), and the Human Protein Atlas (HPA) to validate our results. Results Through an extensive examination of four prominent databases, a total of 21 original articles encompassing a cohort of 3,378 patients were identified. The findings indicated that a low expression of p27 could lead to shorter overall survival (OS) [hazard ratio (HR) = 0.44, 95% confidence interval (95%CI) = 0.31-0.61, Z = 4.89, p = 0.000] and disease-free survival (DFS) (HR = 0.40, 95%CI = 0.28-0.59, Z = 4.75, p = 0.000). In addition, a low expression of p27 predisposed tumors to the right colon [odds ratio (OR) = 0.61, 95%CI = 0.46-0.82, Z = 3.32, p = 0.001] and limited tumor differentiation (OR = 0.56, 95%CI = 0.41-0.77, Z = 3.62, p = 0.000), but had no effect on TNM staging (OR = 0.80, 95%CI = 0.52-1.22, Z = 1.05, p = 0.295), lymph node metastasis (OR = 0.90, 95%CI = 0.25-3.28, Z = 0.16, p = 0.876), and tumor size (OR = 0.94, 95%CI = 0.54-1.65, Z = 0.21, p = 0.835). The results from GEPIA and UALCAN showed that p27 had no effect on TNM staging, lymph node metastasis, DFS, and OS; moreover, there was no expression difference between tumor tissues and normal tissues. The findings from the HPA indicated that there was lower expression of p27 in tumor tissues compared with normal tissues. Conclusion Although inconsistent results were reached with the bioinformatics analysis from this meta-analysis, it was confirmed that a low expression of p27 can adversely affect the prognosis of patients with CRC and make a meaningful impact on a part of the clinicopathological features in the meta-analysis with abundant data. In the future, predicting the prognosis of patients with CRC and guiding treatment might emerge as a significant objective.
Collapse
Affiliation(s)
- Jing Zou
- Department of Radiology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Dong Wang
- Department of Stomach and Intestine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Gaoping Yin
- Department of Stomach and Intestine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Kexiang Lu
- Department of Stomach and Intestine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Kaibin Chang
- Department of Stomach and Intestine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - He Li
- Department of Stomach and Intestine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
7
|
Gu Z, Zou L, Pan X, Yu Y, Liu Y, Zhang Z, Liu J, Mao S, Zhang J, Guo C, Li W, Geng J, Zhang W, Yao X, Shen B. The role and mechanism of NAT10-mediated ac4C modification in tumor development and progression. MedComm (Beijing) 2024; 5:e70026. [PMID: 39640362 PMCID: PMC11617596 DOI: 10.1002/mco2.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024] Open
Abstract
RNA modification has emerged as a crucial area of research in epigenetics, significantly influencing tumor biology by regulating RNA metabolism. N-acetyltransferase 10 (NAT10)-mediated N4-acetylcytidine (ac4C) modification, the sole known acetylation in eukaryotic RNA, influences cancer pathogenesis and progression. NAT10 is the only writer of ac4C and catalyzes acetyl transfer on targeted RNA, and ac4C helps to improve the stability and translational efficiency of ac4C-modified RNA. NAT10 is highly expressed and associated with poor prognosis in pan-cancers. Based on its molecular mechanism and biological functions, ac4C is a central factor in tumorigenesis, tumor progression, drug resistance, and tumor immune escape. Despite the increasing focus on ac4C, the specific regulatory mechanisms of ac4C in cancer remain elusive. The present review thoroughly analyzes the current knowledge on NAT10-mediated ac4C modification in cancer, highlighting its broad regulatory influence on targeted gene expression and tumor biology. This review also summarizes the limitations and perspectives of current research on NAT10 and ac4C in cancer, to identify new therapeutic targets and advance cancer treatment strategies.
Collapse
Affiliation(s)
- Zhuoran Gu
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Libin Zou
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Xinjian Pan
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Yang Yu
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Yongqiang Liu
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Zhijin Zhang
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Ji Liu
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Shiyu Mao
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Junfeng Zhang
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Changcheng Guo
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Wei Li
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Jiang Geng
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Wentao Zhang
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Xudong Yao
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
| | - Bing Shen
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghaiChina
- Urologic Cancer InstituteSchool of MedicineTongji UniversityShanghaiChina
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of MedicineTongi UniversityShanahaiChina
| |
Collapse
|
8
|
Hernández-Ramírez LC, Perez-Rivas LG, Theodoropoulou M, Korbonits M. An Update on the Genetic Drivers of Corticotroph Tumorigenesis. Exp Clin Endocrinol Diabetes 2024; 132:678-696. [PMID: 38830604 DOI: 10.1055/a-2337-2265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
The genetic landscape of corticotroph tumours of the pituitary gland has dramatically changed over the last 10 years. Somatic changes in the USP8 gene account for the most common genetic defect in corticotrophinomas, especially in females, while variants in TP53 or ATRX are associated with a subset of aggressive tumours. Germline defects have also been identified in patients with Cushing's disease: some are well-established (MEN1, CDKN1B, DICER1), while others are rare and could represent coincidences. In this review, we summarise the current knowledge on the genetic drivers of corticotroph tumorigenesis, their molecular consequences, and their impact on the clinical presentation and prognosis.
Collapse
Affiliation(s)
- Laura C Hernández-Ramírez
- Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Marily Theodoropoulou
- Medizinische Klinik und Poliklinik IV, LMU Klinikum, LMU München, Munich 80336, Germany
| | - Márta Korbonits
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, UK
| |
Collapse
|
9
|
Li K, Hong Y, Yu Y, Xie Z, Lv D, Wang C, Xie T, Chen H, Chen Z, Zeng J, Zhao S. NAT10 Promotes Prostate Cancer Growth and Metastasis by Acetylating mRNAs of HMGA1 and KRT8. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310131. [PMID: 38922788 PMCID: PMC11348116 DOI: 10.1002/advs.202310131] [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: 12/22/2023] [Revised: 05/22/2024] [Indexed: 06/28/2024]
Abstract
N4-acetylcytidine (ac4C) is essential for the development and migration of tumor cells. According to earlier research, N-acetyltransferase 10 (NAT10) can increase messenger RNAs (mRNAs) stability by catalyzing the synthesis of ac4C. However, little is known about NAT10 expression and its role in the acetylation modifications in prostate cancer (PCa). Thus, the biological function of NAT10 in PCa is investigated in this study. Compared to paraneoplastic tissues, the expression of NAT10 is significantly higher in PCa. The NAT10 expression is strongly correlated with the pathological grade, clinical stage, Gleason score, T-stage, and N-stage of PCa. NAT10 has the ability to advance the cell cycle and the epithelial-mesenchymal transition (EMT), both of which raise the malignancy of tumor cells. Mechanistically, NAT10 enhance the stability of high mobility group AT-hook 1 (HMGA1) by acetylating its mRNA, thereby promoting cell cycle progression to improve cell proliferation. In addition, NAT10 improve the stability of Keratin 8 (KRT8) by acetylating its mRNA, which promotes the progression of EMT to improve cell migration. This findings provide a potential prognostic or therapeutic target for PCa.
Collapse
Affiliation(s)
- Kang‐Jing Li
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
- Department of UrologyAffiliated Qingyuan HospitalGuangzhou Medical UniversityQingyuan People's HospitalQingyuan511518China
| | - Yaying Hong
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Yu‐Zhong Yu
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Zhiyue Xie
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Dao‐Jun Lv
- Department of UrologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Chong Wang
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Tao Xie
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Hong Chen
- Luoyang Key Laboratory of Organic Functional MoleculesCollege of Food and DrugLuoyang Normal UniversityLuoyangHenan471934P. R. China
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNY11439USA
| | - Jianwen Zeng
- Department of UrologyAffiliated Qingyuan HospitalGuangzhou Medical UniversityQingyuan People's HospitalQingyuan511518China
| | - Shan‐Chao Zhao
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
- Department of UrologyThe Fifth Affiliated HospitalSouthern Medical UniversityGuangzhou510900China
- Department of UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhou510500China
| |
Collapse
|
10
|
Zhang X, Liu M, Wang Z, Wang P, Kong L, Wu J, Wu W, Ma L, Jiang S, Ren W, Du L, Ma W, Liu X. A review of the botany, phytochemistry, pharmacology, synthetic biology and comprehensive utilization of Silybum marianum. Front Pharmacol 2024; 15:1417655. [PMID: 39055491 PMCID: PMC11269164 DOI: 10.3389/fphar.2024.1417655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
Silybum marianum (L.) Gaertn, a herbaceous plant with a long history in traditional medicine for the treatment of hepatobiliary diseases, particularly in Europe, which has attracted attention for its remarkable therapeutic effect. This review systematically summarizes the research progress in the botany, phytochemistry, pharmacology, comprehensive utilization and synthetic biology of S. marianum. Up to now, more than 20 types of flavonolignan components have been isolated from S. marianum. In addition, the rearch on fatty acids and triterpenoids is also constantly improving. Among them, silybin is the most active compound in flavonolignans components. Its pharmacological effects in vivo and in vitro include anti-inflammatory, antioxidant, anti-tumour, hypoglycaemic, neuroprotective and immunoregulatory properties. The use of coniferyl alcohol and taxifolin as substrates to produce silybin and isosilybin under the action of enzyme catalysis is the commonly used biosynthetic pathway of silymarin, which provides support for a comprehensive analysis of the synthetic pathway of silymarin. In addition to medicinal use, the extracts of plants also have broad application prospects in the production of food, healthcare products, cosmetics and other aspects. In addition, the chemical composition, pharmacological mechanism and synthetic biology of S. marianum need to be further studied, which is very important for its clinical efficacy and resource development.
Collapse
Affiliation(s)
- Xiaozhuang Zhang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Meiqi Liu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhen Wang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Panpan Wang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lingyang Kong
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jianhao Wu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Wu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lengleng Ma
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shan Jiang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Weichao Ren
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Likun Du
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Ma
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiubo Liu
- College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi, China
| |
Collapse
|
11
|
Hamar J, Cnaani A, Kültz D. Effects of CRISPR/Cas9 targeting of the myo-inositol biosynthesis pathway on hyper-osmotic tolerance of tilapia cells. Genomics 2024; 116:110833. [PMID: 38518899 DOI: 10.1016/j.ygeno.2024.110833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Myo-inositol is an important compatible osmolyte in vertebrates. This osmolyte is produced by the myo-inositol biosynthesis (MIB) pathway composed of myo-inositol phosphate synthase and inositol monophosphatase. These enzymes are among the highest upregulated proteins in tissues and cell cultures from teleost fish exposed to hyperosmotic conditions indicating high importance of this pathway for tolerating this type of stress. CRISPR/Cas9 gene editing of tilapia cells produced knockout lines of MIB enzymes and control genes. Metabolic activity decreased significantly for MIB KO lines in hyperosmotic media. Trends of faster growth of the MIB knockout lines in isosmotic media and faster decline of MIB knockout lines in hyperosmotic media were also observed. These results indicate a decline in metabolic fitness but only moderate effects on cell survival when tilapia cells with disrupted MIB genes are exposed to hyperosmolality. Therefore MIB genes are required for full osmotolerance of tilapia cells.
Collapse
Affiliation(s)
- Jens Hamar
- Department of Animal Sciences & Genome Center, University of California Davis, Meyer Hall, One Shields Avenue, Davis, CA 95616, USA
| | - Avner Cnaani
- Department of Poultry and Aquaculture, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, P.O. Box 15159, Rishon LeZion 7528809, Israel
| | - Dietmar Kültz
- Department of Animal Sciences & Genome Center, University of California Davis, Meyer Hall, One Shields Avenue, Davis, CA 95616, USA.
| |
Collapse
|
12
|
Ramírez-Rentería C, Hernández-Ramírez LC. Genetic diagnosis in acromegaly and gigantism: From research to clinical practice. Best Pract Res Clin Endocrinol Metab 2024; 38:101892. [PMID: 38521632 DOI: 10.1016/j.beem.2024.101892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2024]
Abstract
It is usually considered that only 5% of all pituitary neuroendocrine tumours are due to inheritable causes. Since this estimate was reported, however, multiple genetic defects driving syndromic and nonsyndromic somatotrophinomas have been unveiled. This heterogeneous genetic background results in overlapping phenotypes of GH excess. Genetic tests should be part of the approach to patients with acromegaly and gigantism because they can refine the clinical diagnoses, opening the possibility to tailor the clinical conduct to each patient. Even more, genetic testing and clinical screening of at-risk individuals have a positive impact on disease outcomes, by allowing for the timely detection and treatment of somatotrophinomas at early stages. Future research should focus on determining the actual frequency of novel genetic drivers of somatotrophinomas in the general population, developing up-to-date disease-specific multi-gene panels for clinical use, and finding strategies to improve access to modern genetic testing worldwide.
Collapse
Affiliation(s)
- Claudia Ramírez-Rentería
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Laura C Hernández-Ramírez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México, e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
13
|
Kim Y, Park WH, Suh DH, Kim K, No JH, Kim YB. Anticancer Effects of BRD4 Inhibitor in Epithelial Ovarian Cancer. Cancers (Basel) 2024; 16:959. [PMID: 38473320 DOI: 10.3390/cancers16050959] [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: 01/15/2024] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Efforts have been made to develop bromodomain inhibitors as cancer treatments. Sub-pathways, particularly in ovarian cancer, affected by bromodomain-containing protein (BRD) remain unclear. This study verified the antitumor effects of a new drug that can overcome OPT-0139-chemoresistance to treat ovarian cancer. A mouse xenograft model of human ovarian cancer cells, SKOV3 and OVCAR3, was used in this study. Cell viability and proliferation were assessed using MTT and ATP assays. Cell cycle arrest and apoptosis were determined using flow cytometry. BRD4 and c-Myc expression and apoptosis-related molecules were detected using RT-PCR and real-time PCR and Western blot. We confirmed the OPT-0139 effect and mechanism of action in epithelial ovarian cancer. OPT-0139 significantly reduced cell viability and proliferation and induced apoptosis and cell cycle arrest. In the mouse xenograft model, significant changes in tumor growth, volume, weight, and BRD4-related gene expression were observed, suggesting the antitumor effects of BRD4 inhibitors. Combination therapy with cisplatin promoted apoptosis and suppressed tumor growth in vitro and in vivo. Our results suggest OPT-0139, a BRD4 inhibitor, as a promising anticancer drug for the treatment of ovarian cancer by inhibiting cell proliferation, decreasing cell viability, arresting cell cycle, and inducing apoptosis.
Collapse
Affiliation(s)
- Yeorae Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
| | - Wook-Ha Park
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
| | - Dong-Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Jongno-gu, Seoul 03080, Republic of Korea
| | - Kidong Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Jongno-gu, Seoul 03080, Republic of Korea
| | - Jae-Hong No
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Jongno-gu, Seoul 03080, Republic of Korea
| | - Yong-Beom Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Jongno-gu, Seoul 03080, Republic of Korea
| |
Collapse
|
14
|
Pessino G, Scotti C, Maggi M, Immuno-Hub Consortium. Hepatocellular Carcinoma: Old and Emerging Therapeutic Targets. Cancers (Basel) 2024; 16:901. [PMID: 38473265 DOI: 10.3390/cancers16050901] [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: 01/31/2024] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Liver cancer, predominantly hepatocellular carcinoma (HCC), globally ranks sixth in incidence and third in cancer-related deaths. HCC risk factors include non-viral hepatitis, alcohol abuse, environmental exposures, and genetic factors. No specific genetic alterations are unequivocally linked to HCC tumorigenesis. Current standard therapies include surgical options, systemic chemotherapy, and kinase inhibitors, like sorafenib and regorafenib. Immunotherapy, targeting immune checkpoints, represents a promising avenue. FDA-approved checkpoint inhibitors, such as atezolizumab and pembrolizumab, show efficacy, and combination therapies enhance clinical responses. Despite this, the treatment of hepatocellular carcinoma (HCC) remains a challenge, as the complex tumor ecosystem and the immunosuppressive microenvironment associated with it hamper the efficacy of the available therapeutic approaches. This review explores current and advanced approaches to treat HCC, considering both known and new potential targets, especially derived from proteomic analysis, which is today considered as the most promising approach. Exploring novel strategies, this review discusses antibody drug conjugates (ADCs), chimeric antigen receptor T-cell therapy (CAR-T), and engineered antibodies. It then reports a systematic analysis of the main ligand/receptor pairs and molecular pathways reported to be overexpressed in tumor cells, highlighting their potential and limitations. Finally, it discusses TGFβ, one of the most promising targets of the HCC microenvironment.
Collapse
Affiliation(s)
- Greta Pessino
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Claudia Scotti
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Maristella Maggi
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Immuno-Hub Consortium
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
15
|
Jeong J, Lee J, Talaia G, Kim W, Song J, Hong J, Yoo K, Gonzalez DG, Athonvarangkul D, Shin J, Dann P, Haberman AM, Kim LK, Ferguson SM, Choi J, Wysolmerski J. Intracellular calcium links milk stasis to lysosome-dependent cell death during early mammary gland involution. Cell Mol Life Sci 2024; 81:29. [PMID: 38212474 PMCID: PMC10784359 DOI: 10.1007/s00018-023-05044-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 10/17/2023] [Accepted: 11/07/2023] [Indexed: 01/13/2024]
Abstract
Involution of the mammary gland after lactation is a dramatic example of coordinated cell death. Weaning causes distension of the alveolar structures due to the accumulation of milk, which, in turn, activates STAT3 and initiates a caspase-independent but lysosome-dependent cell death (LDCD) pathway. Although the importance of STAT3 and LDCD in early mammary involution is well established, it has not been entirely clear how milk stasis activates STAT3. In this report, we demonstrate that protein levels of the PMCA2 calcium pump are significantly downregulated within 2-4 h of experimental milk stasis. Reductions in PMCA2 expression correlate with an increase in cytoplasmic calcium in vivo as measured by multiphoton intravital imaging of GCaMP6f fluorescence. These events occur concomitant with the appearance of nuclear pSTAT3 expression but prior to significant activation of LDCD or its previously implicated mediators such as LIF, IL6, and TGFβ3, all of which appear to be upregulated by increased intracellular calcium. We further demonstrate that increased intracellular calcium activates STAT3 by inducing degradation of its negative regulator, SOCS3. We also observed that milk stasis, loss of PMCA2 expression and increased intracellular calcium levels activate TFEB, an important regulator of lysosome biogenesis through a process involving inhibition of CDK4/6 and cell cycle progression. In summary, these data suggest that intracellular calcium serves as an important proximal biochemical signal linking milk stasis to STAT3 activation, increased lysosomal biogenesis, and lysosome-mediated cell death.
Collapse
Affiliation(s)
- Jaekwang Jeong
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Jongwon Lee
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Gabriel Talaia
- Departments of Cell Biology and of Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Wonnam Kim
- Division of Phamacology, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Junho Song
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Juhyeon Hong
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kwangmin Yoo
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - David G Gonzalez
- Department of Genetics, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Diana Athonvarangkul
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jaehun Shin
- Integrated Science Engineering Division, Underwood International College, Yonsei University, Seoul, Republic of Korea
| | - Pamela Dann
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Ann M Haberman
- Departments of Immunobiology and Laboratory Medicine, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Lark Kyun Kim
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06230, Republic of Korea
| | - Shawn M Ferguson
- Departments of Cell Biology and of Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - John Wysolmerski
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
16
|
Adetutu A, Aborisade AB, Ogunsina FA, Adegbola PI, Olaniyi TD. Ginger mitigated the health risks associated with arsenic-contamination of rats feed via inflammatory and apoptosis regulation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 269:115768. [PMID: 38064790 DOI: 10.1016/j.ecoenv.2023.115768] [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: 09/09/2023] [Revised: 11/12/2023] [Accepted: 11/27/2023] [Indexed: 01/12/2024]
Abstract
Millions of people around the world are inadvertently exposed to arsenic through drinking water and food. However, food spices possess antioxidants and anti-inflammatory potentials. Therefore, this study evaluated the protective potentials of Zingiber officinale (ginger) against the toxic effects of arsenic in male Wistar rats. Thirty-six Wistar rats were assigned into 6 groups (n = 6); group A1 and A2 (control), group B1 and B2 were fed with arsenic-contaminated feed (3.45x10-3 mg/kg), group C1 and C2 were feed with arsenic-contaminated feed (3.45x10-3 mg) supplemented with ginger respectively for 12 and 24 weeks. The blood, bone marrow, and liver of rats were harvested and prepared for various analyses. Micronucleus and Comet analysis were performed for the genotoxicity assessment every 4 weeks. Activities of AST, ALT, GGT, and SOD, and the concentration of GSH, MDA, protein carbonyl, protein thiol, and total protein, were measured by spectrophotometric methods. Quantification of IL-10, 1 L-1β, TNF-α, TGF-β NF-Ƙβ, and 8-oxodeoxyguanosine was done by ELISA method while Bax, Bcl2, and Erk 1/2 were quantified by immuno-histochemical staining. mRNA expression of cyclin D1 was quantified using qRT-PCR. Statistical analysis was performed with SPSS and statistical significance was accepted when p<0.05. Result showed significant (p<0.05) decrease in the haemoglobin concentration, red blood cell, lymphocyte counts, tail DNA and MnPCE of rats fed arsenic-contaminated feed compared with control. The supplementation with ginger significantly reduced serum activities of AST and GGT (p<0.05). Ginger supplementation also lowered the arsenic indued increases in liver MDA, protein carbonyl and 8-OXdG levels. Ginger restores to near normal the histological changes due to arsenic exposure. In the arsenic-exposed group, liver IL-10, IL-1β and TNF-α decreased significantly (p<0.05) at week 24 whereas, NF-Ƙβ and TGF-β increased significantly (p 0.05) at weeks 12 and 24 and TNF-α, Bcl2 at week 24. mRNA expression of cyclin D1 was significantly (p<0.05) downregulated in the arsenic and ginger-supplemented groups. This study showed that long-term consumption of arsenic resulted in immunosuppression, anaemia and activated anti-apoptotic process that was mitigated due to ginger supplementation.
Collapse
Affiliation(s)
- Adewale Adetutu
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Abiodun Bukunmi Aborisade
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Faith Ayotunde Ogunsina
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Peter Ifeoluwa Adegbola
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria; Department of Biochemistry and Forensic Science, First Technical University, Ibadan, Nigeria.
| | - Temitope Deborah Olaniyi
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| |
Collapse
|
17
|
Lye J, Delaney DS, Leith FK, Sardesai VS, McLenachan S, Chen FK, Atlas MD, Wong EYM. Recent Therapeutic Progress and Future Perspectives for the Treatment of Hearing Loss. Biomedicines 2023; 11:3347. [PMID: 38137568 PMCID: PMC10741758 DOI: 10.3390/biomedicines11123347] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Up to 1.5 billion people worldwide suffer from various forms of hearing loss, with an additional 1.1 billion people at risk from various insults such as increased consumption of recreational noise-emitting devices and ageing. The most common type of hearing impairment is sensorineural hearing loss caused by the degeneration or malfunction of cochlear hair cells or spiral ganglion nerves in the inner ear. There is currently no cure for hearing loss. However, emerging frontier technologies such as gene, drug or cell-based therapies offer hope for an effective cure. In this review, we discuss the current therapeutic progress for the treatment of hearing loss. We describe and evaluate the major therapeutic approaches being applied to hearing loss and summarize the key trials and studies.
Collapse
Affiliation(s)
- Joey Lye
- Hearing Therapeutics, Ear Science Institute Australia, Nedlands, WA 6009, Australia; (J.L.); (D.S.D.); (F.K.L.); (V.S.S.); (M.D.A.)
- Centre for Ear Sciences, Medical School, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Derek S. Delaney
- Hearing Therapeutics, Ear Science Institute Australia, Nedlands, WA 6009, Australia; (J.L.); (D.S.D.); (F.K.L.); (V.S.S.); (M.D.A.)
- Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
| | - Fiona K. Leith
- Hearing Therapeutics, Ear Science Institute Australia, Nedlands, WA 6009, Australia; (J.L.); (D.S.D.); (F.K.L.); (V.S.S.); (M.D.A.)
- Centre for Ear Sciences, Medical School, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Varda S. Sardesai
- Hearing Therapeutics, Ear Science Institute Australia, Nedlands, WA 6009, Australia; (J.L.); (D.S.D.); (F.K.L.); (V.S.S.); (M.D.A.)
| | - Samuel McLenachan
- Ocular Tissue Engineering Laboratory, Lions Eye Institute, Nedlands, WA 6009, Australia; (S.M.); (F.K.C.)
- Centre for Ophthalmology and Visual Sciences, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Fred K. Chen
- Ocular Tissue Engineering Laboratory, Lions Eye Institute, Nedlands, WA 6009, Australia; (S.M.); (F.K.C.)
- Centre for Ophthalmology and Visual Sciences, The University of Western Australia, Nedlands, WA 6009, Australia
- Vitroretinal Surgery, Royal Perth Hospital, Perth, WA 6000, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC 3002, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Marcus D. Atlas
- Hearing Therapeutics, Ear Science Institute Australia, Nedlands, WA 6009, Australia; (J.L.); (D.S.D.); (F.K.L.); (V.S.S.); (M.D.A.)
- Centre for Ear Sciences, Medical School, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Elaine Y. M. Wong
- Hearing Therapeutics, Ear Science Institute Australia, Nedlands, WA 6009, Australia; (J.L.); (D.S.D.); (F.K.L.); (V.S.S.); (M.D.A.)
- Centre for Ear Sciences, Medical School, The University of Western Australia, Nedlands, WA 6009, Australia
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia
| |
Collapse
|
18
|
McMahon RA, D'Souza C, Neeson PJ, Siva S. Innate immunity: Looking beyond T-cells in radiation and immunotherapy combinations. Neoplasia 2023; 46:100940. [PMID: 37913654 PMCID: PMC10637988 DOI: 10.1016/j.neo.2023.100940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023]
Abstract
Radiation therapy is an established and effective anti-cancer treatment modality. Extensive pre-clinical experimentation has demonstrated that the pro-inflammatory properties of irradiation may be synergistic with checkpoint immunotherapy. Radiation induces double-stranded DNA breaks (dsDNA). Sensing of the dsDNA activates the cGAS/STING pathway, producing Type 1 interferons essential to recruiting antigen-presenting cells (APCs). Radiation promotes cytotoxic CD8 T-cell recruitment by releasing tumour-associated antigens captured and cross-presented by surveying antigen-presenting cells. Radiation-induced vascular normalisation may further promote T-cell trafficking and drug delivery. Radiation is also immunosuppressive. Recruitment of regulatory T cells (Tregs) and innate cells such as myeloid-derived suppressive cells (m-MDSCs) all counteract the immunostimulatory properties of radiation. Many innate immune cell types operate at the interface of the adaptive immune response. Innate immune cells, such as m-MDSCs, can exert their immunosuppressive effects by expressing immune checkpoints such as PD-L1, further highlighting the potential of combined radiation and checkpoint immunotherapy. Several early-phase clinical studies investigating the combination of radiation and immunotherapy have been disappointing. A greater appreciation of radiotherapy's impact on the innate immune system is essential to optimise radioimmunotherapy combinations. This review will summarise the impact of radiotherapy on crucial cells of the innate immune system and vital immunosuppressive cytokines.
Collapse
Affiliation(s)
- R A McMahon
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.
| | - C D'Souza
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia; Cancer Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - P J Neeson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia; Cancer Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - S Siva
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia
| |
Collapse
|
19
|
Hüneke H, Langeheine M, Rode K, Jung K, Pilatz A, Fietz D, Kliesch S, Brehm R. Effects of a Sertoli cell-specific knockout of Connexin43 on maturation and proliferation of postnatal Sertoli cells. Differentiation 2023; 134:31-51. [PMID: 37839230 DOI: 10.1016/j.diff.2023.09.002] [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: 05/04/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023]
Abstract
Adult male Sertoli cell-specific Connexin43 knockout mice (SCCx43KO) exhibit higher Sertoli cell (SC) numbers per seminiferous tubule compared to their wild type (WT) littermates. Thus, deletion of this testicular gap junction protein seems to affect the proliferative potential and differentiation of "younger" SC. Although SC have so far mostly been characterised as postmitotic cells that cease to divide and become an adult, terminally differentiated cell population at around puberty, there is rising evidence that there exist exceptions from this for a very long time accepted paradigm. Aim of this study was to investigate postnatal SC development and to figure out underlying causes for observed higher SC numbers in adult KO mice. Therefore, the amount of SC mitotic figures was compared, resulting in slightly more and prolonged detection of SC mitotic figures in KO mice compared to WT. SC counting per tubular cross section revealed significantly different time curves, and comparing proliferation rates using Bromodesoxyuridine and Sox9 showed higher proliferation rates in 8-day old KO mice. SC proliferation was further investigated by Ki67 immunohistochemistry. SC in KO mice displayed a delayed initiation of cell-cycle-inhibitor p27Kip1 synthesis and prolonged synthesis of the phosphorylated tumour suppressor pRb and proliferation marker Ki67. Thus, the higher SC numbers in adult male SCCx43KO mice may arise due to two different reasons: Firstly, in prepubertal KO mice, the proliferation rate of SC was higher. Secondly, there were differences in their ability to cease proliferation as shown by the delayed initiation of p27Kip1 synthesis and the prolonged production of phosphorylated pRb and Ki67. Immunohistochemical results indicating a prolonged period of SC proliferation in SCCx43KO were confirmed by detection of proliferating SC in 17-days-old KO mice. In conclusion, deletion of the testicular gap junction protein Cx43 might prevent normal SC maturation and might even alter also the proliferation potential of adult SC.
Collapse
Affiliation(s)
- Hanna Hüneke
- Institute of Anatomy, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Marion Langeheine
- Institute of Anatomy, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Kristina Rode
- Institute of Anatomy, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Klaus Jung
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Adrian Pilatz
- Department of Urology, Pediatric Urology and Andrology, Justus Liebig University Giessen, Giessen, Germany
| | - Daniela Fietz
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Giessen, Germany
| | - Sabine Kliesch
- Centre of Andrology and Reproductive Medicine, University of Muenster, Muenster, Germany
| | - Ralph Brehm
- Institute of Anatomy, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany.
| |
Collapse
|
20
|
Lim Y, Tamayo-Orrego L, Schmid E, Tarnauskaite Z, Kochenova OV, Gruar R, Muramatsu S, Lynch L, Schlie AV, Carroll PL, Chistol G, Reijns MAM, Kanemaki MT, Jackson AP, Walter JC. In silico protein interaction screening uncovers DONSON's role in replication initiation. Science 2023; 381:eadi3448. [PMID: 37590370 PMCID: PMC10801813 DOI: 10.1126/science.adi3448] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/08/2023] [Indexed: 08/19/2023]
Abstract
CDC45-MCM2-7-GINS (CMG) helicase assembly is the central event in eukaryotic replication initiation. In yeast, a multi-subunit "pre-loading complex" (pre-LC) accompanies GINS to chromatin-bound MCM2-7, leading to CMG formation. Here, we report that DONSON, a metazoan protein mutated in microcephalic primordial dwarfism, is required for CMG assembly in vertebrates. Using AlphaFold to screen for protein-protein interactions followed by experimental validation, we show that DONSON scaffolds a vertebrate pre-LC containing GINS, TOPBP1, and DNA pol ε. Our evidence suggests that DONSON docks the pre-LC onto MCM2-7, delivering GINS to its binding site in CMG. A patient-derived DONSON mutation compromises CMG assembly and recapitulates microcephalic dwarfism in mice. These results unify our understanding of eukaryotic replication initiation, implicate defective CMG assembly in microcephalic dwarfism, and illustrate how in silico protein-protein interaction screening accelerates mechanistic discovery.
Collapse
Affiliation(s)
- Yang Lim
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Blavatnik Institute; Boston, MA 02115, USA
| | - Lukas Tamayo-Orrego
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh; Edinburgh, EH4 2XU, UK
| | - Ernst Schmid
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Blavatnik Institute; Boston, MA 02115, USA
| | - Zygimante Tarnauskaite
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh; Edinburgh, EH4 2XU, UK
| | - Olga V. Kochenova
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Blavatnik Institute; Boston, MA 02115, USA
- Howard Hughes Medical Institute; Boston, MA 02115, USA
| | - Rhian Gruar
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Blavatnik Institute; Boston, MA 02115, USA
| | - Sachiko Muramatsu
- Department of Chromosome Science, National Institute of Genetics, Research Organization of Information and Systems (ROIS); Mishima, Shizuoka 411-8540, Japan
| | - Luke Lynch
- Biochemistry Department, Stanford School of Medicine; Stanford, CA 94305, USA
| | - Aitana Verdu Schlie
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh; Edinburgh, EH4 2XU, UK
| | - Paula L. Carroll
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh; Edinburgh, EH4 2XU, UK
| | - Gheorghe Chistol
- Chemical and Systems Biology Department, Stanford School of Medicine; Stanford, CA 94305, USA
| | - Martin A. M. Reijns
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh; Edinburgh, EH4 2XU, UK
| | - Masato T. Kanemaki
- Department of Chromosome Science, National Institute of Genetics, Research Organization of Information and Systems (ROIS); Mishima, Shizuoka 411-8540, Japan
- Graduate Institute for Advanced Studies, SOKENDAI; Mishima, Shizuoka 411-8540, Japan
- Department of Biological Science, The University of Tokyo; Tokyo 113-0033, Japan
| | - Andrew P. Jackson
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh; Edinburgh, EH4 2XU, UK
| | - Johannes C. Walter
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Blavatnik Institute; Boston, MA 02115, USA
- Howard Hughes Medical Institute; Boston, MA 02115, USA
| |
Collapse
|
21
|
Rowland RJ, Heath R, Maskell D, Thompson RF, Ranson NA, Blaza JN, Endicott JA, Noble MEM, Salamina M. Cryo-EM structure of SKP1-SKP2-CKS1 in complex with CDK2-cyclin A-p27KIP1. Sci Rep 2023; 13:10718. [PMID: 37400515 PMCID: PMC10318019 DOI: 10.1038/s41598-023-37609-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/24/2023] [Indexed: 07/05/2023] Open
Abstract
p27KIP1 (cyclin-dependent kinase inhibitor 1B, p27) is a member of the CIP/KIP family of CDK (cyclin dependent kinase) regulators that inhibit cell cycle CDKs. p27 phosphorylation by CDK1/2, signals its recruitment to the SCFSKP2 (S-phase kinase associated protein 1 (SKP1)-cullin-SKP2) E3 ubiquitin ligase complex for proteasomal degradation. The nature of p27 binding to SKP2 and CKS1 was revealed by the SKP1-SKP2-CKS1-p27 phosphopeptide crystal structure. Subsequently, a model for the hexameric CDK2-cyclin A-CKS1-p27-SKP1-SKP2 complex was proposed by overlaying an independently determined CDK2-cyclin A-p27 structure. Here we describe the experimentally determined structure of the isolated CDK2-cyclin A-CKS1-p27-SKP1-SKP2 complex at 3.4 Å global resolution using cryogenic electron microscopy. This structure supports previous analysis in which p27 was found to be structurally dynamic, transitioning from disordered to nascent secondary structure on target binding. We employed 3D variability analysis to further explore the conformational space of the hexameric complex and uncovered a previously unidentified hinge motion centred on CKS1. This flexibility gives rise to open and closed conformations of the hexameric complex that we propose may contribute to p27 regulation by facilitating recognition with SCFSKP2. This 3D variability analysis further informed particle subtraction and local refinement approaches to enhance the local resolution of the complex.
Collapse
Affiliation(s)
- Rhianna J Rowland
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Richard Heath
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Daniel Maskell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Rebecca F Thompson
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
- Life Sciences Electron Microscopy, Thermo Fisher Scientific, Leeds, UK
| | - Neil A Ranson
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - James N Blaza
- Department of Chemistry, York Structural Biology Laboratory and York Biomedical Research Institute, University of York, Heslington, YO10 5DD, York, UK
| | - Jane A Endicott
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Martin E M Noble
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Marco Salamina
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
- Evotec (UK) Ltd., Milton, Abingdon, OX14 4RZ, UK.
| |
Collapse
|
22
|
Park Y, Choo SP, Jung GS, Kim S, Lee MJ, Im W, Park H, Lee I, Lee JH, Cho S, Choi YS. Formononetin Inhibits Progression of Endometriosis via Regulation of p27, pSTAT3, and Progesterone Receptor: In Vitro and In Vivo Studies. Nutrients 2023; 15:3001. [PMID: 37447325 DOI: 10.3390/nu15133001] [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: 05/24/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
OBJECTIVES Formononetin is one of the phytoestrogens that functions like a selective estrogen receptor modulator (SERM). In this study, we evaluated the effects of formononetin on endometriosis progression in vitro and in vivo. MATERIALS AND METHODS After pathological confirmation, 10 eutopic and ectopic endometria were collected from patients with endometriosis. Ten eutopic endometria samples were collected from patients who did not have endometriosis. To determine the cytotoxic dose and therapeutic dose of formononetin, the concentration of 70% of the cells that survived after formononetin administration was estimated using a Cell counting kit-8 (CCK 8) assay. Western blot analysis was used to determine the relative expression levels of BAX, p53, pAKT, ERK, pERK, p27, and pSTAT3 in the eutopic endometria without endometriosis, eutopic endometria with endometriosis, and ectopic endometria with endometriosis as the formononetin concentration was increased. We confirmed the effect of formononetin on apoptosis and migration in endometriosis using fluorescence-activated cell sorting (FACS) and wound healing assays, respectively. A mouse model of endometriosis was prepared using a non-surgical method, as previously described. The mice were intraperitoneally administered formononetin for four weeks after dividing them into control, low-dose formononetin (40 mg/kg/day) treatment, and high-dose (80 mg/kg/day) formononetin treatment groups. All the mice were euthanized after formononetin treatment. Endometriotic lesions were retrieved and confirmed using hematoxylin and eosin (H&E) staining. Immunohistochemical (IHC) staining of p27 was performed. RESULTS We set the maximum concentration of formononetin administration to 80 μM through the CCK8 assay. Based on formononetin concentration, the expression levels of BAX, p53, pAKT, ERK, pERK, p27, and pSTAT3 proteins were measured using Western blot analysis (N = 4 per group). The expression level of pERK, p27, and pSTAT3 in eutopic endometrium with endometriosis tended to decrease with increasing formononetin concentration, and a significant decrease was noted at 80 μM. The expression of p27 in ectopic endometrium with endometriosis was also significantly decreased at 80 μM of formononetin. FACS analysis revealed that formononetin did not significantly affect apoptosis. In the wound healing assay, formononetin treatment revealed a more significant decrease in the proliferation of the eutopic endometrium in patients with endometriosis than in the eutopic endometrium without endometriosis. Relative expression of sex hormone receptors decreased with increasing formononetin doses. Although no significant differences were observed in the ER, PR-A, ERβ/ERα, and PR-B/PR-A, significant down-regulation of PR-B expression was noted after formononetin treatment at 80 μM. In the in vivo study, endometriotic lesions in the formononetin-treated group significantly decreased compared to those in the control group. The relative expression of p27 using IHC was highest in the control group and lowest in the high-dose formononetin treatment group. CONCLUSIONS Formononetin treatment was shown to inhibit the proliferation of eutopic and ectopic endometria in patients with endometriosis through the regulation of p27, pSTAT3, and PR-B. In an endometriosis mouse model, formononetin treatment significantly reduced the number of endometriotic lesions with decreased p27 expression. The results of this study suggest that formononetin may be used as a non-hormonal treatment option for endometriosis.
Collapse
Affiliation(s)
- Yunjeong Park
- Department of Obstetrics and Gynecology, Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
| | - Sung Pil Choo
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Gee Soo Jung
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
- Department of Medical Device Engineering and Management, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
| | - Sehee Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
- Department of Medical Device Engineering and Management, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
| | - Min Jung Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Wooseok Im
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyemin Park
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
| | - Inha Lee
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
| | - Jae Hoon Lee
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sihyun Cho
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Republic of Korea
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Young Sik Choi
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
23
|
Jeong J, Lee J, Talaia G, Kim W, Song J, Hong J, Yoo K, Gonzalez D, Athonvarangkul D, Shin J, Dann P, Haberman A, Kim LK, Ferguson S, Choi J, Wysolmerski J. Intracellular Calcium links Milk Stasis to Lysosome Dependent Cell Death by Activating a TGFβ3/TFEB/STAT3 Pathway Early during Mammary Gland Involution. RESEARCH SQUARE 2023:rs.3.rs-3030763. [PMID: 37398309 PMCID: PMC10312953 DOI: 10.21203/rs.3.rs-3030763/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Involution of the mammary gland after lactation is a dramatic example of coordinated cell death. Weaning causes distension of the alveolar structures due to the accumulation of milk, which, in turn, activates STAT3 and initiates a caspase-independent but lysosome-dependent cell death (LDCD) pathway. Although the importance of STAT3 and LDCD in early mammary involution is well established, it has not been entirely clear how milk stasis activates STAT3. In this report, we demonstrate that protein levels of the PMCA2 calcium pump are significantly downregulated within 2-4 hours of experimental milk stasis. Reductions in PMCA2 expression correlate with an increase in cytoplasmic calcium in vivo as measured by multiphoton intravital imaging of GCaMP6f fluorescence. These events occur concomitant with the appearance of nuclear pSTAT3 expression but prior to significant activation of LDCD or its previously implicated mediators such as LIF, IL6 and TGFβ3, all of which appear to be upregulated by increased intracellular calcium. We also observed that milk stasis, loss of PMCA2 expression and increased intracellular calcium levels activate TFEB, an important regulator of lysosome biogenesis. This is the result of increased TGFβ signaling and inhibition of cell cycle progression. Finally, we demonstrate that increased intracellular calcium activates STAT3 by inducing degradation of its negative regulator, SOCS3, a process which also appears to be mediated by TGFβ signaling. In summary, these data suggest that intracellular calcium serves as an important proximal biochemical signal linking milk stasis to STAT3 activation, increased lysosomal biogenesis, and lysosome-mediated cell death.
Collapse
Affiliation(s)
- Jaekwang Jeong
- Yale School of Medicine: Yale University School of Medicine
| | | | - Gabriel Talaia
- Yale School of Medicine: Yale University School of Medicine
| | | | | | | | | | - David Gonzalez
- Yale School of Medicine: Yale University School of Medicine
| | | | | | - Pamela Dann
- Yale School of Medicine: Yale University School of Medicine
| | - Ann Haberman
- Yale School of Medicine: Yale University School of Medicine
| | | | - Shawn Ferguson
- Yale School of Medicine: Yale University School of Medicine
| | | | | |
Collapse
|
24
|
Ford K, Munson BP, Fong SH, Panwala R, Chu WK, Rainaldi J, Plongthongkum N, Arunachalam V, Kostrowicki J, Meluzzi D, Kreisberg JF, Jensen-Pergakes K, VanArsdale T, Paul T, Tamayo P, Zhang K, Bienkowska J, Mali P, Ideker T. Multimodal perturbation analyses of cyclin-dependent kinases reveal a network of synthetic lethalities associated with cell-cycle regulation and transcriptional regulation. Sci Rep 2023; 13:7678. [PMID: 37169829 PMCID: PMC10175263 DOI: 10.1038/s41598-023-33329-2] [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] [Received: 11/15/2022] [Accepted: 04/11/2023] [Indexed: 05/13/2023] Open
Abstract
Cell-cycle control is accomplished by cyclin-dependent kinases (CDKs), motivating extensive research into CDK targeting small-molecule drugs as cancer therapeutics. Here we use combinatorial CRISPR/Cas9 perturbations to uncover an extensive network of functional interdependencies among CDKs and related factors, identifying 43 synthetic-lethal and 12 synergistic interactions. We dissect CDK perturbations using single-cell RNAseq, for which we develop a novel computational framework to precisely quantify cell-cycle effects and diverse cell states orchestrated by specific CDKs. While pairwise disruption of CDK4/6 is synthetic-lethal, only CDK6 is required for normal cell-cycle progression and transcriptional activation. Multiple CDKs (CDK1/7/9/12) are synthetic-lethal in combination with PRMT5, independent of cell-cycle control. In-depth analysis of mRNA expression and splicing patterns provides multiple lines of evidence that the CDK-PRMT5 dependency is due to aberrant transcriptional regulation resulting in premature termination. These inter-dependencies translate to drug-drug synergies, with therapeutic implications in cancer and other diseases.
Collapse
Affiliation(s)
- Kyle Ford
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Brenton P Munson
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Samson H Fong
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Rebecca Panwala
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Wai Keung Chu
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Joseph Rainaldi
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
- Biomedical Sciences Program, University of California San Diego, La Jolla, CA, 92093, USA
| | - Nongluk Plongthongkum
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | | | | | - Dario Meluzzi
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jason F Kreisberg
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | | | - Todd VanArsdale
- Pfizer Inc, 10555 Science Center Drive, San Diego, CA, 92121, USA
| | - Thomas Paul
- Pfizer Inc, 10555 Science Center Drive, San Diego, CA, 92121, USA
| | - Pablo Tamayo
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Kun Zhang
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | | | - Prashant Mali
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Trey Ideker
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA.
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
25
|
Ku AT, Shankavaram U, Trostel SY, Zhang H, Sater HA, Harmon SA, Carrabba NV, Liu Y, Wood BJ, Pinto PA, Choyke PL, Stoyanova R, Davicioni E, Pollack A, Turkbey B, Sowalsky AG, Citrin DE. Radiogenomic profiling of prostate tumors prior to external beam radiotherapy converges on a transcriptomic signature of TGF-β activity driving tumor recurrence. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.01.23288883. [PMID: 37205576 PMCID: PMC10187349 DOI: 10.1101/2023.05.01.23288883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Background Patients with localized prostate cancer have historically been assigned to clinical risk groups based on local disease extent, serum prostate specific antigen (PSA), and tumor grade. Clinical risk grouping is used to determine the intensity of treatment with external beam radiotherapy (EBRT) and androgen deprivation therapy (ADT), yet a substantial proportion of patients with intermediate and high risk localized prostate cancer will develop biochemical recurrence (BCR) and require salvage therapy. Prospective identification of patients destined to experience BCR would allow treatment intensification or selection of alternative therapeutic strategies. Methods Twenty-nine individuals with intermediate or high risk prostate cancer were prospectively recruited to a clinical trial designed to profile the molecular and imaging features of prostate cancer in patients undergoing EBRT and ADT. Whole transcriptome cDNA microarray and whole exome sequencing were performed on pretreatment targeted biopsy of prostate tumors (n=60). All patients underwent pretreatment and 6-month post EBRT multiparametric MRI (mpMRI), and were followed with serial PSA to assess presence or absence of BCR. Genes differentially expressed in the tumor of patients with and without BCR were investigated using pathways analysis tools and were similarly explored in alternative datasets. Differential gene expression and predicted pathway activation were evaluated in relation to tumor response on mpMRI and tumor genomic profile. A novel TGF-β gene signature was developed in the discovery dataset and applied to a validation dataset. Findings Baseline MRI lesion volume and PTEN/TP53 status in prostate tumor biopsies correlated with the activation state of TGF-β signaling measured using pathway analysis. All three measures correlated with the risk of BCR after definitive RT. A prostate cancer-specific TGF-β signature discriminated between patients that experienced BCR vs. those that did not. The signature retained prognostic utility in an independent cohort. Interpretation TGF-β activity is a dominant feature of intermediate-to-unfavorable risk prostate tumors prone to biochemical failure after EBRT with ADT. TGF-β activity may serve as a prognostic biomarker independent of existing risk factors and clinical decision-making criteria. Funding This research was supported by the Prostate Cancer Foundation, the Department of Defense Congressionally Directed Medical Research Program, National Cancer Institute, and the Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research.
Collapse
Affiliation(s)
- Anson T. Ku
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, MD, USA
| | - Uma Shankavaram
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Shana Y. Trostel
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, MD, USA
| | - Hong Zhang
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Houssein A. Sater
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, MD, USA
| | | | - Nicole V. Carrabba
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, MD, USA
| | - Yang Liu
- Veracyte, Inc., South San Francisco, CA, USA
| | - Bradford J. Wood
- Center for Interventional Oncology, NIH Clinical Center, Bethesda, MD, USA
| | - Peter A. Pinto
- Urologic Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Peter L. Choyke
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD, USA
| | - Radka Stoyanova
- Department of Radiation Oncology, University of Miami, Miami, FL, USA
| | | | - Alan Pollack
- Department of Radiation Oncology, University of Miami, Miami, FL, USA
| | - Baris Turkbey
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD, USA
| | - Adam G. Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, MD, USA
| | - Deborah E. Citrin
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
26
|
Hatamzade Esfahani N, Day AS. The Role of TGF-β, Activin and Follistatin in Inflammatory Bowel Disease. GASTROINTESTINAL DISORDERS 2023; 5:167-186. [DOI: 10.3390/gidisord5020015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/10/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an immune-mediated inflammatory condition predominantly affecting the gastrointestinal (GI) tract. An increasing prevalence of IBD has been observed globally. The pathogenesis of IBD includes a complex interplay between the intestinal microbiome, diet, genetic factors and immune responses. The consequent imbalance of inflammatory mediators ultimately leads to intestinal mucosal damage and defective repair. Growth factors, given their specific roles in maintaining the homeostasis and integrity of the intestinal epithelium, are of particular interest in the setting of IBD. Furthermore, direct targeting of growth factor signalling pathways involved in the regeneration of the damaged epithelium and the regulation of inflammation could be considered as therapeutic options for individuals with IBD. Several members of the transforming growth factor (TGF)-β superfamily, particularly TGF-β, activin and follistatin, are key candidates as they exhibit various roles in inflammatory processes and contribute to maintenance and homeostasis in the GI tract. This article aimed firstly to review the events involved in the pathogenesis of IBD with particular emphasis on TGF-β, activin and follistatin and secondly to outline the potential role of therapeutic manipulation of these pathways.
Collapse
Affiliation(s)
| | - Andrew S. Day
- Paediatric Department, University of Otago Christchurch, Christchurch 8140, New Zealand
| |
Collapse
|
27
|
The CMG helicase and cancer: a tumor "engine" and weakness with missing mutations. Oncogene 2023; 42:473-490. [PMID: 36522488 PMCID: PMC9948756 DOI: 10.1038/s41388-022-02572-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/01/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
The replicative Cdc45-MCM-GINS (CMG) helicase is a large protein complex that functions in the DNA melting and unwinding steps as a component of replisomes during DNA replication in mammalian cells. Although the CMG performs this important role in cell growth, the CMG is not a simple bystander in cell cycle events. Components of the CMG, specifically the MCM precursors, are also involved in maintaining genomic stability by regulating DNA replication fork speeds, facilitating recovery from replicative stresses, and preventing consequential DNA damage. Given these important functions, MCM/CMG complexes are highly regulated by growth factors such as TGF-ß1 and by signaling factors such as Myc, Cyclin E, and the retinoblastoma protein. Mismanagement of MCM/CMG complexes when these signaling mediators are deregulated, and in the absence of the tumor suppressor protein p53, leads to increased genomic instability and is a contributor to tumorigenic transformation and tumor heterogeneity. The goal of this review is to provide insight into the mechanisms and dynamics by which the CMG is regulated during its assembly and activation in mammalian genomes, and how errors in CMG regulation due to oncogenic changes promote tumorigenesis. Finally, and most importantly, we highlight the emerging understanding of the CMG helicase as an exploitable vulnerability and novel target for therapeutic intervention in cancer.
Collapse
|
28
|
Zhang T, He M, Zhang J, Tong Y, Chen T, Wang C, Pan W, Xiao Z. Mechanisms of primordial follicle activation and new pregnancy opportunity for premature ovarian failure patients. Front Physiol 2023; 14:1113684. [PMID: 36926197 PMCID: PMC10011087 DOI: 10.3389/fphys.2023.1113684] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Primordial follicles are the starting point of follicular development and the basic functional unit of female reproduction. Primordial follicles are formed around birth, and most of the primordial follicles then enter a dormant state. Since primordial follicles are limited in number and can't be renewed, dormant primordial follicles cannot be reversed once they enter the growing state. Thus, the orderly occurrence of primordial follicles selective activation directly affects the rate of follicle consumption and thus determines the length of female reproductive lifespan. Studies have found that appropriately inhibiting the activation rate of primordial follicles can effectively slow down the rate of follicle consumption, maintain fertility and delay ovarian aging. Based on the known mechanisms of primordial follicle activation, primordial follicle in vitro activation (IVA) technique has been clinically developed. IVA can help patients with premature ovarian failure, middle-aged infertile women, or infertile women due to gynecological surgery treatment to solve infertility problems. The study of the mechanism of selective activation of primordial follicles can contribute to the development of more efficient and safe IVA techniques. In this paper, recent mechanisms of primordial follicle activation and its clinical application are reviewed.
Collapse
Affiliation(s)
- Tuo Zhang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.,Prenatal Diagnosis Center in Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China.,Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Meina He
- College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.,Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jingjing Zhang
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yuntong Tong
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Tengxiang Chen
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.,College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China.,Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Wei Pan
- Prenatal Diagnosis Center in Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ziwen Xiao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
29
|
Zheng Y, Yu Z, Li Y, Zhong S, Sun Y, Sun L, Zheng X, Qi X, Zhang S. Alcohol extracts of Chinese bayberry branch induce S-phase arrest and apoptosis in HepG2 cells. Food Sci Nutr 2023; 11:493-503. [PMID: 36655066 PMCID: PMC9834848 DOI: 10.1002/fsn3.3080] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 08/31/2022] [Accepted: 09/15/2022] [Indexed: 01/21/2023] Open
Abstract
The alcohol extracts of Chinese bayberry (Myrica rubra) branches (MRBE) are rich in flavonoids which have a variety of medicinal benefits, but their effects on human HepG2 were unknown. In this study, the effects of MRBE on HepG2 cell growth and its potential for inhibiting cancer were explored. The results displayed that MRBE inhibited HepG2 proliferation both by arresting cells in S phase and promoting apoptosis. Quantitative reverse-transcription PCR (qRT-PCR), western blotting, and immunofluorescence showed that MRBE induced S-phase arrest by upregulating p21, which in turn downregulated cyclin and cyclin-dependent kinase messenger RNA (mRNA) and protein. Apoptosis was induced by blocking the expression of BCL-2 and suppression of the Raf/ERK1 signaling pathways. These results indicated that MRBE may have the potential for treatment of human liver cancer, highlighting novel approaches for developing new pharmacological tools for the treatment of this deadly type cancer. Meanwhile, it provides a new direction for the medicinal added values of Chinese bayberry, which helped to broaden the diversified development of its industry chain.
Collapse
Affiliation(s)
- Yuanyuan Zheng
- Institute of HorticultureZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Zheping Yu
- Institute of HorticultureZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Yougui Li
- Institute of Sericultural and TeaZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Shi Zhong
- Institute of Sericultural and TeaZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Yuqing Sun
- Institute of Sericultural and TeaZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Li Sun
- Institute of HorticultureZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Xiliang Zheng
- Institute of HorticultureZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Xingjiang Qi
- Institute of HorticultureZhejiang Academy of Agricultural SciencesHangzhouChina
- Xianghu LabHangzhouChina
| | - Shuwen Zhang
- Institute of HorticultureZhejiang Academy of Agricultural SciencesHangzhouChina
| |
Collapse
|
30
|
Giri S, Park GH, Choi JS, Ma E, Chun KS, Joo SH. MS-5, a Naphthalene Derivative, Induces Apoptosis in Human Pancreatic Cancer BxPC-3 Cells by Modulating Reactive Oxygen Species. Biomol Ther (Seoul) 2023; 31:68-72. [PMID: 36380602 PMCID: PMC9810442 DOI: 10.4062/biomolther.2022.127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/07/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
Pancreatic cancer is one of the most fatal cancers with a poor prognosis. Standard chemotherapies have proven largely ineffective because of their toxicity and the development of resistance. Therefore, there is an urgent need to develop novel therapies. In this study, we investigated the antitumor activity of MS-5, a naphthalene derivative, on BxPC-3, a human pancreatic cancer cell line. We observed that MS-5 was cytotoxic to BxPC-3 cells, as well as inhibited the growth of cells in a concentration- and time- dependent manner. Flow cytometry analysis revealed that the percentage of annexin V-positive cells increased after MS-5 treatment. We also observed cleavage of caspases and poly (ADP-ribose) polymerase, and downregulation of Bcl-xL protein. Flow cytometry analysis of intracellular levels of reactive oxygen species (ROS) and mitochondrial superoxide suggested that MS-5 induced the generation of mitochondrial superoxide while lowering the overall intracellular ROS levels. Thus, MS-5 may be potential candidate for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Suman Giri
- Department of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Gyu Hwan Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Joon-Seok Choi
- Department of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Eunsook Ma
- Department of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea,Corresponding Authors E-mail: (Joo SH), (Chun KS), Tel: +82-53-850-3614 (Joo SH), +82-53-580-6647 (Chun KS), Fax: +82-53-359-6729 (Joo SH), +82-53-580-6645 (Chun KS)
| | - Sang Hoon Joo
- Department of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea,Corresponding Authors E-mail: (Joo SH), (Chun KS), Tel: +82-53-850-3614 (Joo SH), +82-53-580-6647 (Chun KS), Fax: +82-53-359-6729 (Joo SH), +82-53-580-6645 (Chun KS)
| |
Collapse
|
31
|
Sohn HA, Kang M, Ha H, Yeom YI, Park KC, Lee DC. R-PTP-κ Inhibits Contact-Dependent Cell Growth by Suppressing E2F Activity. Biomedicines 2022; 10:biomedicines10123199. [PMID: 36551956 PMCID: PMC9775357 DOI: 10.3390/biomedicines10123199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Density-dependent regulation of cell growth is presumed to be caused by cell-cell contact, but the underlying molecular mechanism is not yet clearly defined. Here, we report that receptor-type protein tyrosine phosphatase-kappa (R-PTP-κ) is an important regulator of cell contact-dependent growth inhibition. R-PTP-κ expression increased in proportion to cell density. siRNA-mediated R-PTP-κ downregulation led to the loss of cell contact-mediated growth inhibition, whereas its upregulation reduced anchorage-independent cell growth in soft agar as well as tumor growth in nude mice. Expression profiling and luciferase reporter system-mediated signaling pathway analysis revealed that R-PTP-κ induced under cell contact conditions distinctly suppressed E2F activity. Among the structural domains of R-PTP-κ, the cytoplasmic domain containing the tandemly repeated PTP motif acts as a potent downregulator of the E2F pathway. Specifically, R-PTP-κ suppressed CDK2 activity through the induction of p21Cip1/WAF-1 and p27Kip1, resulting in cell cycle arrest at the G1 phase. In transcriptome-based public datasets generated from four different tumor types, R-PTP-κ expression was negatively correlated with the expression pattern and prognostic value of two known E2F1 target genes (CCNE1 and CDC25A). Therefore, our results indicate that the R-PTP-κ-E2F axis plays a crucial role in cell growth-inhibitory signaling arising from cell-cell contact conditions.
Collapse
Affiliation(s)
- Hyun Ahm Sohn
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Minho Kang
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Hyunjung Ha
- Department of Biochemistry, School of Life Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Young Il Yeom
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Kyung Chan Park
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
- Correspondence: (K.C.P.); (D.C.L.); Tel.: +82-42-879-8115 (K.C.P.); +82-42-879-8153 (D.C.L.)
| | - Dong Chul Lee
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
- Correspondence: (K.C.P.); (D.C.L.); Tel.: +82-42-879-8115 (K.C.P.); +82-42-879-8153 (D.C.L.)
| |
Collapse
|
32
|
Yuan P, Qin HY, Wei JY, Chen G, Li X. Proteomics reveals the potential mechanism of Tanshinone IIA in promoting the Ex Vivo expansion of human bone marrow mesenchymal stem cells. Regen Ther 2022; 21:560-573. [DOI: 10.1016/j.reth.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
|
33
|
Kim S, Leong A, Kim M, Yang HW. CDK4/6 initiates Rb inactivation and CDK2 activity coordinates cell-cycle commitment and G1/S transition. Sci Rep 2022; 12:16810. [PMID: 36207346 PMCID: PMC9546874 DOI: 10.1038/s41598-022-20769-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 09/19/2022] [Indexed: 02/04/2023] Open
Abstract
External signaling controls cell-cycle entry until cells irreversibly commit to the cell cycle to ensure faithful DNA replication. This process is tightly regulated by cyclin-dependent kinases (CDKs) and the retinoblastoma protein (Rb). Here, using live-cell sensors for CDK4/6 and CDK2 activities, we propose that CDK4/6 initiates Rb inactivation and CDK2 activation, which coordinates the timing of cell-cycle commitment and sequential G1/S transition. Our data show that CDK4/6 activation induces Rb inactivation and thereby E2F activation, driving a gradual increase in CDK2 activity. We found that rapid CDK4/6 inhibition can reverse cell-cycle entry until CDK2 activity reaches to high levels. This suggests that high CDK2 activity is required to initiate CDK2-Rb positive feedback and CDK4/6-indpendent cell-cycle progression. Since CDK2 activation also facilitates initiation of DNA replication, the timing of CDK2-Rb positive feedback is coupled with the G1/S transition. Our experiments, which acutely increased CDK2 activity by cyclin E1 overexpression, indicate that cells commit to the cell cycle before triggering DNA replication. Together, our data suggest that CDK4/6 inactivates Rb to begin E2F and CDK2 activation, and high CDK2 activity is necessary and sufficient to generate a bistable switch for Rb phosphorylation before DNA replication. These findings highlight how cells initiate the cell cycle and subsequently commit to the cell cycle before the G1/S transition.
Collapse
Affiliation(s)
- Sungsoo Kim
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - Alessandra Leong
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - Minah Kim
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA.
| | - Hee Won Yang
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
34
|
Beeken J, Kessels S, Rigo JM, Alpizar YA, Nguyen L, Brône B. p27 kip1 Modulates the Morphology and Phagocytic Activity of Microglia. Int J Mol Sci 2022; 23:10432. [PMID: 36142366 PMCID: PMC9499407 DOI: 10.3390/ijms231810432] [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: 07/18/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
Abstract
p27kip1 is a multifunctional protein that promotes cell cycle exit by blocking the activity of cyclin/cyclin-dependent kinase complexes as well as migration and motility via signaling pathways that converge on the actin and microtubule cytoskeleton. Despite the broad characterization of p27kip1 function in neural cells, little is known about its relevance in microglia. Here, we studied the role of p27kip1 in microglia using a combination of in vitro and in situ approaches. While the loss of p27kip1 did not affect microglial density in the cerebral cortex, it altered their morphological complexity in situ. However, despite the presence of p27kip1 in microglial processes, as shown by immunofluorescence in cultured cells, loss of p27kip1 did not change microglial process motility and extension after applying laser-induced brain damage in cortical brain slices. Primary microglia lacking p27kip1 showed increased phagocytic uptake of synaptosomes, while a cell cycle dead variant negatively affected phagocytosis. These findings indicate that p27kip1 plays specific roles in microglia.
Collapse
Affiliation(s)
- Jolien Beeken
- UHasselt, Hasselt University, BIOMED, 3500 Hasselt, Belgium
- Laboratory of Molecular Regulation of Neurogenesis, GIGA-Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, C.H.U. Sar-Tilman, 4000 Liège, Belgium
| | - Sofie Kessels
- UHasselt, Hasselt University, BIOMED, 3500 Hasselt, Belgium
| | | | | | - Laurent Nguyen
- Laboratory of Molecular Regulation of Neurogenesis, GIGA-Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, C.H.U. Sar-Tilman, 4000 Liège, Belgium
| | - Bert Brône
- UHasselt, Hasselt University, BIOMED, 3500 Hasselt, Belgium
| |
Collapse
|
35
|
Schirripa A, Sexl V, Kollmann K. Cyclin-dependent kinase inhibitors in malignant hematopoiesis. Front Oncol 2022; 12:916682. [PMID: 36033505 PMCID: PMC9403899 DOI: 10.3389/fonc.2022.916682] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
The cell-cycle is a tightly orchestrated process where sequential steps guarantee cellular growth linked to a correct DNA replication. The entire cell division is controlled by cyclin-dependent kinases (CDKs). CDK activation is balanced by the activating cyclins and CDK inhibitors whose correct expression, accumulation and degradation schedule the time-flow through the cell cycle phases. Dysregulation of the cell cycle regulatory proteins causes the loss of a controlled cell division and is inevitably linked to neoplastic transformation. Due to their function as cell-cycle brakes, CDK inhibitors are considered as tumor suppressors. The CDK inhibitors p16INK4a and p15INK4b are among the most frequently altered genes in cancer, including hematopoietic malignancies. Aberrant cell cycle regulation in hematopoietic stem cells (HSCs) bears severe consequences on hematopoiesis and provokes hematological disorders with a broad array of symptoms. In this review, we focus on the importance and prevalence of deregulated CDK inhibitors in hematological malignancies.
Collapse
|
36
|
Kotschenreuther K, Yan S, Kofler DM. Migration and homeostasis of regulatory T cells in rheumatoid arthritis. Front Immunol 2022; 13:947636. [PMID: 36016949 PMCID: PMC9398455 DOI: 10.3389/fimmu.2022.947636] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/20/2022] [Indexed: 12/17/2022] Open
Abstract
Regulatory T (Treg) cells are garnering increased attention in research related to autoimmune diseases, including rheumatoid arthritis (RA). They play an essential role in the maintenance of immune homeostasis by restricting effector T cell activity. Reduced functions and frequencies of Treg cells contribute to the pathogenesis of RA, a common autoimmune disease which leads to systemic inflammation and erosive joint destruction. Treg cells from patients with RA are characterized by impaired functions and by an altered phenotype. They show increased plasticity towards Th17 cells and a reduced suppressive capacity. Besides the suppressive function of Treg cells, their effectiveness is determined by their ability to migrate into inflamed tissues. In the past years, new mechanisms involved in Treg cell migration have been identified. One example of such a mechanism is the phosphorylation of vasodilator-stimulated phosphoprotein (VASP). Efficient migration of Treg cells requires the presence of VASP. IL-6, a cytokine which is abundantly present in the peripheral blood and in the synovial tissue of RA patients, induces posttranslational modifications of VASP. Recently, it has been shown in mice with collagen-induced arthritis (CIA) that this IL-6 mediated posttranslational modification leads to reduced Treg cell trafficking. Another protein which facilitates Treg cell migration is G-protein-signaling modulator 2 (GPSM2). It modulates G-protein coupled receptor functioning, thereby altering the cellular activity initiated by cell surface receptors in response to extracellular signals. The almost complete lack of GPSM2 in Treg cells from RA patients contributes to their reduced ability to migrate towards inflammatory sites. In this review article, we highlight the newly identified mechanisms of Treg cell migration and review the current knowledge about impaired Treg cell homeostasis in RA.
Collapse
Affiliation(s)
- Konstantin Kotschenreuther
- Laboratory of Molecular Immunology, Division of Rheumatology and Clinical Immunology, Department I of Internal Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Shuaifeng Yan
- Laboratory of Molecular Immunology, Division of Rheumatology and Clinical Immunology, Department I of Internal Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - David M. Kofler
- Laboratory of Molecular Immunology, Division of Rheumatology and Clinical Immunology, Department I of Internal Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- *Correspondence: David M. Kofler,
| |
Collapse
|
37
|
From cyclins to CDKIs: Cell cycle regulation of skeletal muscle stem cell quiescence and activation. Exp Cell Res 2022; 420:113275. [PMID: 35931143 DOI: 10.1016/j.yexcr.2022.113275] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/12/2022] [Accepted: 07/03/2022] [Indexed: 11/22/2022]
Abstract
After extensive proliferation during development, the adult skeletal muscle cells remain outside the cell cycle, either as post-mitotic myofibers or as quiescent muscle stem cells (MuSCs). Despite its terminally differentiated state, adult skeletal muscle has a remarkable regeneration potential, driven by MuSCs. Upon injury, MuSC quiescence is reversed to support tissue growth and repair and it is re-established after the completion of muscle regeneration. The distinct cell cycle states and transitions observed in the different myogenic populations are orchestrated by elements of the cell cycle machinery. This consists of i) complexes of cyclins and Cyclin-Dependent Kinases (CDKs) that ensure cell cycle progression and ii) their negative regulators, the Cyclin-Dependent Kinase Inhibitors (CDKIs). In this review we discuss the roles of these factors in developmental and adult myogenesis, with a focus on CDKIs that have emerging roles in stem cell functions.
Collapse
|
38
|
Chiu CW, Hsieh CY, Yang CH, Tsai JH, Huang SY, Sheu JR. Yohimbine, an α2-Adrenoceptor Antagonist, Suppresses PDGF-BB-Stimulated Vascular Smooth Muscle Cell Proliferation by Downregulating the PLCγ1 Signaling Pathway. Int J Mol Sci 2022; 23:ijms23148049. [PMID: 35887391 PMCID: PMC9324260 DOI: 10.3390/ijms23148049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/22/2022] Open
Abstract
Yohimbine (YOH) has antiproliferative effects against breast cancer and pancreatic cancer; however, its effects on vascular proliferative diseases such as atherosclerosis remain unknown. Accordingly, we investigated the inhibitory mechanisms of YOH in vascular smooth muscle cells (VSMCs) stimulated by platelet-derived growth factor (PDGF)-BB, a major mitogenic factor in vascular diseases. YOH (5–20 μM) suppressed PDGF-BB-stimulated a mouse VSMC line (MOVAS-1 cell) proliferation without inducing cytotoxicity. YOH also exhibited antimigratory effects and downregulated matrix metalloproteinase-2 and -9 expression in PDGF-BB-stimulated MOVAS-1 cells. It also promoted cell cycle arrest in the initial gap/first gap phase by upregulating p27Kip1 and p53 expression and reducing cyclin-dependent kinase 2 and proliferating cell nuclear antigen expression. We noted phospholipase C-γ1 (PLCγ1) but not ERK1/2, AKT, or p38 kinase phosphorylation attenuation in YOH-modulated PDGF-BB-propagated signaling pathways in the MOVAS-1 cells. Furthermore, YOH still inhibited PDGF-BB-induced cell proliferation and PLCγ1 phosphorylation in MOVAS-1 cells with α2B-adrenergic receptor knockdown. YOH (5 and 10 mg/kg) substantially suppressed neointimal hyperplasia in mice subjected to CCA ligation for 21 days. Overall, our results reveal that YOH attenuates PDGF-BB-stimulated VSMC proliferation and migration by downregulating a α2B-adrenergic receptor–independent PLCγ1 pathway and reduces neointimal formation in vivo. Therefore, YOH has potential for repurposing for treating atherosclerosis and other vascular proliferative diseases.
Collapse
Affiliation(s)
- Chih-Wei Chiu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Cheng-Ying Hsieh
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-H.Y.); (J.-H.T.)
| | - Chih-Hao Yang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-H.Y.); (J.-H.T.)
| | - Jie-Heng Tsai
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-H.Y.); (J.-H.T.)
| | - Shih-Yi Huang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110, Taiwan
- Center for Reproductive Medicine & Sciences, Taipei Medical University Hospital, Taipei 110, Taiwan
- Correspondence: (S.-Y.H.); (J.-R.S.); Tel.: +886-2-2736-1661 (ext. 6543) (S.-Y.H.); +886-2-2736-1661 (ext. 3199) (J.-R.S.)
| | - Joen-Rong Sheu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-H.Y.); (J.-H.T.)
- Correspondence: (S.-Y.H.); (J.-R.S.); Tel.: +886-2-2736-1661 (ext. 6543) (S.-Y.H.); +886-2-2736-1661 (ext. 3199) (J.-R.S.)
| |
Collapse
|
39
|
Ng JWK, Ong EHQ, Tucker-Kellogg L, Tucker-Kellogg G. Deep learning for de-convolution of Smad2 versus Smad3 binding sites. BMC Genomics 2022; 23:525. [PMID: 35858839 PMCID: PMC9297549 DOI: 10.1186/s12864-022-08565-x] [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: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 11/10/2022] Open
Abstract
Background The transforming growth factor beta-1 (TGF β-1) cytokine exerts both pro-tumor and anti-tumor effects in carcinogenesis. An increasing body of literature suggests that TGF β-1 signaling outcome is partially dependent on the regulatory targets of downstream receptor-regulated Smad (R-Smad) proteins Smad2 and Smad3. However, the lack of Smad-specific antibodies for ChIP-seq hinders convenient identification of Smad-specific binding sites. Results In this study, we use localization and affinity purification (LAP) tags to identify Smad-specific binding sites in a cancer cell line. Using ChIP-seq data obtained from LAP-tagged Smad proteins, we develop a convolutional neural network with long-short term memory (CNN-LSTM) as a deep learning approach to classify a pool of Smad-bound sites as being Smad2- or Smad3-bound. Our data showed that this approach is able to accurately classify Smad2- versus Smad3-bound sites. We use our model to dissect the role of each R-Smad in the progression of breast cancer using a previously published dataset. Conclusions Our results suggests that deep learning approaches can be used to dissect binding site specificity of closely related transcription factors. Supplementary Information The online version contains supplementary material available at (10.1186/s12864-022-08565-x).
Collapse
Affiliation(s)
- Jeremy W K Ng
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Esther H Q Ong
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Lisa Tucker-Kellogg
- Cancer and Stem Cell Biology, and Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore.
| | - Greg Tucker-Kellogg
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore. .,Computational Biology Programme, Faculty of Science, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
40
|
Seabrook A, Wijewardene A, De Sousa S, Wong T, Sheriff N, Gill AJ, Iyer R, Field M, Luxford C, Clifton-Bligh R, McCormack A, Tucker K. MEN4, the MEN1 Mimicker: A Case Series of three Phenotypically Heterogenous Patients With Unique CDKN1B Mutations. J Clin Endocrinol Metab 2022; 107:2339-2349. [PMID: 35323929 PMCID: PMC9282358 DOI: 10.1210/clinem/dgac162] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Indexed: 12/29/2022]
Abstract
CONTEXT Germline CDKN1B pathogenic variants result in multiple endocrine neoplasia type 4 (MEN4), an autosomal dominant hereditary tumor syndrome variably associated with primary hyperparathyroidism, pituitary adenoma, and duodenopancreatic neuroendocrine tumors. OBJECTIVE To report the phenotype of 3 unrelated cases each with a unique germline CDKN1B variant (of which 2 are novel) and compare these cases with those described in the current literature. DESIGN/METHODS Three case studies, including clinical presentation, germline, and tumor genetic analysis and family history. SETTING Two tertiary University Hospitals in Sydney, New South Wales, and 1 tertiary University Hospital in Canberra, Australian Capital Territory, Australia. OUTCOME Phenotype of the 3 cases and their kindred; molecular analysis and tumor p27kip1 immunohistochemistry. RESULTS Family A: The proband developed multiglandular primary hyperparathyroidism, a microprolactinoma and a multifocal nonfunctioning duodenopancreatic neuroendocrine tumor. Family B: The proband was diagnosed with primary hyperparathyroidism from a single parathyroid adenoma. Family C: The proband was diagnosed with a nonfunctioning pituitary microadenoma and ectopic Cushing's syndrome from an atypical thymic carcinoid tumor. Germline sequencing in each patient identified a unique variant in CDKN1B, 2 of which are novel (c.179G > A, p.Trp60*; c.475G > A, p.Asp159Asn) and 1 previously reported (c.374_375delCT, p.Ser125*). CONCLUSIONS Germline CDKN1B pathogenic variants cause the syndrome MEN4. The phenotype resulting from the 3 pathogenic variants described in this series highlights the heterogenous nature of this syndrome, ranging from isolated primary hyperparathyroidism to the full spectrum of endocrine manifestations. We report the first described cases of a prolactinoma and an atypical thymic carcinoid tumor in MEN4.
Collapse
Affiliation(s)
- Amanda Seabrook
- Cancer Genetics Laboratory, Kolling Institute, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- The University of Sydney, Sydney, NSW, 2006, Australia
| | - Ayanthi Wijewardene
- Cancer Genetics Laboratory, Kolling Institute, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- The University of Sydney, Sydney, NSW, 2006, Australia
| | - Sunita De Sousa
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, 5000
- South Australian Adult Genetics Unit, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Tang Wong
- The University of New South Wales, Sydney, NSW, 2052, Australia
- The University of Western Sydney, Sydney, NSW, 2560, Australia
- Department of Endocrinology, Prince of Wales Hospital, Sydney, NSW, 2064, Australia
| | - Nisa Sheriff
- Department of Endocrinology, Hornsby Ku-ring-gai Hospital, Sydney, NSW, 2077, Australia
| | - Anthony J Gill
- The University of Sydney, Sydney, NSW, 2006, Australia
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, NSW, 2064, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute, Royal North Shore Hospital, Sydney, NSW, 2064, Australia
| | - Rakesh Iyer
- Calvary Public Hospital, Canberra, ACT, 2617, Australia
| | - Michael Field
- Familial Cancer Service, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
| | - Catherine Luxford
- Cancer Genetics Laboratory, Kolling Institute, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- The University of Sydney, Sydney, NSW, 2006, Australia
| | | | | | - Katherine Tucker
- Correspondence: Katherine Tucker, MBBS, FRACP, AO, Hereditary Cancer Service Nelune Comprehensive Cancer Centre (Bright Building), 64-66 High St, Randwick, NSW, 2031, Australia.
| |
Collapse
|
41
|
Xiang Z, Liqing Y, Qingqing Y, Qiang H, Hongbo C. Retard or exacerbate: Role of long non-coding RNA growth arrest-specific 5 in the fibrosis. Cytokine Growth Factor Rev 2022; 67:89-104. [DOI: 10.1016/j.cytogfr.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/26/2022]
|
42
|
Targeting tumor cell senescence and polyploidy as potential therapeutic strategies. Semin Cancer Biol 2022; 81:37-47. [PMID: 33358748 PMCID: PMC8214633 DOI: 10.1016/j.semcancer.2020.12.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 01/14/2023]
Abstract
Senescence is a unique state of growth arrest that develops in response to a plethora of cellular stresses, including replicative exhaustion, oxidative injury, and genotoxic insults. Senescence has been implicated in the pathogenesis of multiple aging-related pathologies, including cancer. In cancer, senescence plays a dual role, initially acting as a barrier against tumor progression by enforcing a durable growth arrest in premalignant cells, but potentially promoting malignant transformation in neighboring cells through the secretion of pro-tumorigenic drivers. Moreover, senescence is induced in tumor cells upon exposure to a wide variety of conventional and targeted anticancer drugs (termed Therapy-Induced Senescence-TIS), representing a critical contributing factor to therapeutic outcomes. As with replicative or oxidative senescence, TIS manifests as a complex phenotype of macromolecular damage, energetic dysregulation, and altered gene expression. Senescent cells are also frequently polyploid. In vitro studies have suggested that polyploidy may confer upon senescent tumor cells the ability to escape from growth arrest, thereby providing an additional avenue whereby tumor cells escape the lethality of anticancer treatment. Polyploidy in tumor cells is also associated with persistent energy production, chromatin remodeling, self-renewal, stemness and drug resistance - features that are also associated with escape from senescence and conversion to a more malignant phenotype. However, senescent cells are highly heterogenous and can present with variable phenotypes, where polyploidy is one component of a complex reversion process. Lastly, emerging efforts to pharmacologically target polyploid tumor cells might pave the way towards the identification of novel targets for the elimination of senescent tumor cells by the incorporation of senolytic agents into cancer therapeutic strategies.
Collapse
|
43
|
Sager RA, Backe SJ, Ahanin E, Smith G, Nsouli I, Woodford MR, Bratslavsky G, Bourboulia D, Mollapour M. Therapeutic potential of CDK4/6 inhibitors in renal cell carcinoma. Nat Rev Urol 2022; 19:305-320. [PMID: 35264774 PMCID: PMC9306014 DOI: 10.1038/s41585-022-00571-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2022] [Indexed: 12/12/2022]
Abstract
The treatment of advanced and metastatic kidney cancer has entered a golden era with the addition of more therapeutic options, improved survival and new targeted therapies. Tyrosine kinase inhibitors, mammalian target of rapamycin (mTOR) inhibitors and immune checkpoint blockade have all been shown to be promising strategies in the treatment of renal cell carcinoma (RCC). However, little is known about the best therapeutic approach for individual patients with RCC and how to combat therapeutic resistance. Cancers, including RCC, rely on sustained replicative potential. The cyclin-dependent kinases CDK4 and CDK6 are involved in cell-cycle regulation with additional roles in metabolism, immunogenicity and antitumour immune response. Inhibitors of CDK4 and CDK6 are now commonly used as approved and investigative treatments in breast cancer, as well as several other tumours. Furthermore, CDK4/6 inhibitors have been shown to work synergistically with other kinase inhibitors, including mTOR inhibitors, as well as with immune checkpoint inhibitors in preclinical cancer models. The effect of CDK4/6 inhibitors in kidney cancer is relatively understudied compared with other cancers, but the preclinical studies available are promising. Collectively, growing evidence suggests that targeting CDK4 and CDK6 in kidney cancer, alone and in combination with current therapeutics including mTOR and immune checkpoint inhibitors, might have therapeutic benefit and should be further explored.
Collapse
Affiliation(s)
- Rebecca A Sager
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Sarah J Backe
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Elham Ahanin
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Garrett Smith
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Imad Nsouli
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
- Syracuse VA Medical Center, Syracuse, NY, USA
| | - Mark R Woodford
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Gennady Bratslavsky
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Dimitra Bourboulia
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Mehdi Mollapour
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA.
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.
- Syracuse VA Medical Center, Syracuse, NY, USA.
| |
Collapse
|
44
|
Bencivenga D, Stampone E, Vastante A, Barahmeh M, Della Ragione F, Borriello A. An Unanticipated Modulation of Cyclin-Dependent Kinase Inhibitors: The Role of Long Non-Coding RNAs. Cells 2022; 11:cells11081346. [PMID: 35456025 PMCID: PMC9028986 DOI: 10.3390/cells11081346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/13/2022] Open
Abstract
It is now definitively established that a large part of the human genome is transcribed. However, only a scarce percentage of the transcriptome (about 1.2%) consists of RNAs that are translated into proteins, while the large majority of transcripts include a variety of RNA families with different dimensions and functions. Within this heterogeneous RNA world, a significant fraction consists of sequences with a length of more than 200 bases that form the so-called long non-coding RNA family. The functions of long non-coding RNAs range from the regulation of gene transcription to the changes in DNA topology and nucleosome modification and structural organization, to paraspeckle formation and cellular organelles maturation. This review is focused on the role of long non-coding RNAs as regulators of cyclin-dependent kinase inhibitors’ (CDKIs) levels and activities. Cyclin-dependent kinases are enzymes necessary for the tuned progression of the cell division cycle. The control of their activity takes place at various levels. Among these, interaction with CDKIs is a vital mechanism. Through CDKI modulation, long non-coding RNAs implement control over cellular physiology and are associated with numerous pathologies. However, although there are robust data in the literature, the role of long non-coding RNAs in the modulation of CDKIs appears to still be underestimated, as well as their importance in cell proliferation control.
Collapse
|
45
|
Regulation of p27 (Kip1) by Ubiquitin E3 Ligase RNF6. Pharmaceutics 2022; 14:pharmaceutics14040802. [PMID: 35456636 PMCID: PMC9029106 DOI: 10.3390/pharmaceutics14040802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/29/2022] [Accepted: 04/02/2022] [Indexed: 12/10/2022] Open
Abstract
The cyclin-dependent kinase inhibitor p27 (Kip1) is an important regulator of the G1/S checkpoint. It is degraded by the SCF-SKP2 complex in late G1 thereby allowing cells to progress to the S phase. Here we investigated the role of the E3 ubiquitin ligase RNF6 (Ring Finger Protein 6) in cell cycle progression in prostate cancer cells. Our data demonstrate that RNF6 can promote cell cycle progression by reducing the levels of p27. Knockdown of RNF6 led to an increase in the stability of p27 and to the arrest of cells in the G1 phase. RNF6 interacted with p27 via its KIL domain and this interaction was found to be phosphorylation independent. RNF6 enhanced ubiquitination and subsequent degradation of p27 in the early G0/G1 phase of the cell cycle. Knockdown of RNF6 expression by short hairpin RNA led to inhibition of the CDK2/Cyclin E complex thereby reducing phosphorylation of Retinoblastoma protein (Rb) and to a subsequent decrease in cell cycle progression and proliferation. Our data suggest that RNF6 acts as a negative regulator for p27kip1 leading to its proteasome-dependent degradation in the early G0/G1 phase of the cell cycle.
Collapse
|
46
|
Khan RA, Hossain R, Roy P, Jain D, Mohammad Saikat AS, Roy Shuvo AP, Akram M, Elbossaty WF, Khan IN, Painuli S, Semwal P, Rauf A, Islam MT, Khan H. Anticancer effects of acteoside: Mechanistic insights and therapeutic status. Eur J Pharmacol 2022; 916:174699. [PMID: 34919888 DOI: 10.1016/j.ejphar.2021.174699] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/26/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022]
Abstract
Cancer, the uncontrolled proliferation and metastasis of abnormal cells, is a major public health issue worldwide. To date, several natural compounds have been reported with their efficacy in the treatment of different types of cancer. Chemotherapeutic agents are used in cancer treatment and prevention, among other aspects. Acteoside is a phenylethanoid glycoside, first isolated from Verbascum sinuatum, which has demonstrated multiple effects, including antioxidant, anti-epileptic, neuroprotective, anti-inflammatory, antifungal, antihypertensive, and anti-leishmanial properties. This review gathered, analyzed, and summarized the literature on acteoside and its anticancer properties. All the available information about this compound and its role in different types of cancer was collected using different scientific search engines, including PubMed, Scopus, Springer Link, Wiley Online, Web of Science, Scifinder, ScienceDirect, and Google Scholar. Acteoside is found in a variety of plants and has been shown to have anticancer activity in many experimental models through oxidative stress, apoptosis, anti-angiogenesis, anti-invasion, anti-metastasis, synergism with other agents, and anti-proliferative effects through modulation of several pathways. In conclusion, acteoside exhibited potent anticancer activity against different cancer cell lines through modulating several cancer signaling pathways in different non- and pre-clinical experimental models and thus could be a strong candidate for further clinical studies.
Collapse
Affiliation(s)
- Rasel Ahmed Khan
- Pharmacy Discipline, Khulna University, Khulna, 9280, Bangladesh
| | - Rajib Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Dhaka, Bangladesh
| | - Pranta Roy
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430064, Hubei, China
| | - Divya Jain
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan University, Tonk, 304022, India
| | - Abu Saim Mohammad Saikat
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Dhaka, Bangladesh
| | - Anik Prasad Roy Shuvo
- Department of Pharmacy, Southern University Bangladesh, Mehedibag Road, Chattagram, 4000, Bangladesh
| | - Muhammad Akram
- Department of Eastern Medicine, Government College University Faisalabad, 38000, Pakistan
| | | | - Ishaq N Khan
- Institute of Basic Medical Sciences Khyber Medical University, Peshawar, 25100, Pakistan
| | - Sakshi Painuli
- Himalayan Environmental Studies and Conservation Organization (HESCO), Dehradun, 248006, Uttarakhand, India
| | - Prabhakar Semwal
- Department of Life Sciences, Graphic Era Demeed to be University, Dehradun, 248002, Uttarakhand, India
| | - Abdur Rauf
- Department of Chemistry University of Swabi, Swabi, Anbar, 23430, KPK, Pakistan.
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Dhaka, Bangladesh.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| |
Collapse
|
47
|
Hayashi S, Sakata S, Kawamura S, Tokutake Y, Yonekura S. XBP1u Is Involved in C2C12 Myoblast Differentiation via Accelerated Proteasomal Degradation of Id3. Front Physiol 2022; 13:796190. [PMID: 35153829 PMCID: PMC8829448 DOI: 10.3389/fphys.2022.796190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/05/2022] [Indexed: 11/13/2022] Open
Abstract
Myoblast differentiation is an ordered multistep process that includes withdrawal from the cell cycle, elongation, and fusion to form multinucleated myotubes. Id3, a member of the Id family, plays a crucial role in cell cycle exit and differentiation. However, in muscle cells after differentiation induction, the detailed mechanisms that diminish Id3 function and cause the cells to withdraw from the cell cycle are unknown. Induction of myoblast differentiation resulted in decreased expression of Id3 and increased expression of XBP1u, and XBP1u accelerated proteasomal degradation of Id3 in C2C12 cells. The expression levels of the cyclin-dependent kinase inhibitors p21, p27, and p57 were not increased after differentiation induction of XBP1-knockdown C2C12 cells. Moreover, knockdown of Id3 rescued myogenic differentiation of XBP1-knockdown C2C12 cells. Taken together, these findings provide evidence that XBP1u regulates cell cycle exit after myogenic differentiation induction through interactions with Id3. To the best of our knowledge, this is the first report of the involvement of XBP1u in myoblast differentiation. These results indicate that XBP1u may act as a “regulator” of myoblast differentiation under various physiological conditions.
Collapse
Affiliation(s)
- Satoko Hayashi
- Graduate School of Medicine, Science and Technology, Shinshu University, Nagano, Japan
| | - Shotaro Sakata
- Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Shotaro Kawamura
- Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Yukako Tokutake
- Graduate School of Medicine, Science and Technology, Shinshu University, Nagano, Japan
| | - Shinichi Yonekura
- Graduate School of Medicine, Science and Technology, Shinshu University, Nagano, Japan
- Graduate School of Science and Technology, Shinshu University, Nagano, Japan
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
- *Correspondence: Shinichi Yonekura,
| |
Collapse
|
48
|
Endo I, Amatya VJ, Kushitani K, Kambara T, Nakagiri T, Fujii Y, Takeshima Y. Insulin-Like Growth Factor 2 mRNA Binding Protein 3 Promotes Cell Proliferation of Malignant Mesothelioma Cells by Downregulating p27Kip1. Front Oncol 2022; 11:795467. [PMID: 35127504 PMCID: PMC8807558 DOI: 10.3389/fonc.2021.795467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/31/2021] [Indexed: 11/17/2022] Open
Abstract
Malignant mesothelioma is a tumor with a poor prognosis, mainly caused by asbestos exposure and with no adequate treatment yet. To develop future therapeutic targets, we analyzed the microarray dataset GSE 29370 of malignant mesothelioma and reactive mesothelial hyperplasia, downloaded from the Gene Expression Omnibus (GEO) database. We identified insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3) as one of the significantly upregulated genes in malignant mesothelioma. IGF2BP3 functions as an oncoprotein in many human cancers; however, to our knowledge, this is the first study on the biological function of IGF2BP3 in malignant mesothelioma cells. The knockdown of IGF2BP3 in malignant mesothelioma cells resulted in the suppression of cell proliferation with an increase in the proportion of cells in the G1 phase of the cell cycle. Furthermore, knockdown of IGF2BP3 inhibited cell migration and invasion. We focused on the cell cycle assay to investigate the role of IGF2BP3 in cell proliferation in malignant mesothelioma. Among the various proteins involved in cell cycle regulation, the expression of p27 Kip1 (p27) increased significantly upon IGF2BP3 knockdown. Next, p27 siRNA was added to suppress the increased expression of p27. The results showed that p27 knockdown attenuated the effects of IGF2BP3 knockdown on cell proliferation and G1 phase arrest. In conclusion, we found that IGF2BP3 promotes cell proliferation, a critical step in tumorigenesis, by suppressing the expression of p27 in malignant mesothelioma.
Collapse
|
49
|
Ettl T, Schulz D, Bauer RJ. The Renaissance of Cyclin Dependent Kinase Inhibitors. Cancers (Basel) 2022; 14:293. [PMID: 35053461 PMCID: PMC8773807 DOI: 10.3390/cancers14020293] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/29/2021] [Accepted: 01/04/2022] [Indexed: 12/14/2022] Open
Abstract
Cyclin-dependent kinases (CDK) regulate cell cycle progression. During tumor development, altered expression and availability of CDKs strongly contribute to impaired cell proliferation, a hallmark of cancer. In recent years, targeted inhibition of CDKs has shown considerable therapeutic benefit in a variety of tumor entities. Their success is reflected in clinical approvals of specific CDK4/6 inhibitors for breast cancer. This review provides a detailed insight into the molecular mechanisms of CDKs as well as a general overview of CDK inhibition. It also summarizes the latest research approaches and current advances in the treatment of head and neck cancer with CDK inhibitors. Instead of monotherapies, combination therapies with CDK inhibitors may especially provide promising results in tumor therapy. Indeed, recent studies have shown a synergistic effect of CDK inhibition together with chemo- and radio- and immunotherapy in cancer treatment to overcome tumor evasion, which may lead to a renaissance of CDK inhibitors.
Collapse
Affiliation(s)
- Tobias Ettl
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Daniela Schulz
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany;
- Center for Medical Biotechnology, Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Richard Josef Bauer
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany;
- Center for Medical Biotechnology, Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
50
|
Lim AR, Ghajar CM. Thorny ground, rocky soil: Tissue-specific mechanisms of tumor dormancy and relapse. Semin Cancer Biol 2022; 78:104-123. [PMID: 33979673 PMCID: PMC9595433 DOI: 10.1016/j.semcancer.2021.05.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023]
Abstract
Disseminated tumor cells (DTCs) spread systemically yet distinct patterns of metastasis indicate a range of tissue susceptibility to metastatic colonization. Distinctions between permissive and suppressive tissues are still being elucidated at cellular and molecular levels. Although there is a growing appreciation for the role of the microenvironment in regulating metastatic success, we have a limited understanding of how diverse tissues regulate DTC dormancy, the state of reversible quiescence and subsequent awakening thought to contribute to delayed relapse. Several themes of microenvironmental regulation of dormancy are beginning to emerge, including vascular association, co-option of pre-existing niches, metabolic adaptation, and immune evasion, with tissue-specific nuances. Conversely, DTC awakening is often associated with injury or inflammation-induced activation of the stroma, promoting a proliferative environment with DTCs following suit. We review what is known about tissue-specific regulation of tumor dormancy on a tissue-by-tissue basis, profiling major metastatic organs including the bone, lung, brain, liver, and lymph node. An aerial view of the barriers to metastatic growth may reveal common targets and dependencies to inform the therapeutic prevention of relapse.
Collapse
Affiliation(s)
- Andrea R Lim
- Public Health Sciences Division/Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Graduate Program in Molecular and Cellular Biology, University of Washington/Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| | - Cyrus M Ghajar
- Public Health Sciences Division/Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| |
Collapse
|