1
|
Jiang Y, Zhou R, Liao F, Kong G, Zeng J, Wu Y, Li X, Wang B, Qi F, Chen S, Zhu Q, Gu L, Zheng C. Unraveling radiation-induced skeletal muscle damage: Insights from a 3D human skeletal muscle organoid model. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119792. [PMID: 38936620 DOI: 10.1016/j.bbamcr.2024.119792] [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: 03/06/2024] [Revised: 05/28/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Three-dimensional (3D) organoids derived from human pluripotent stem cells (hPSCs) have revolutionized in vitro tissue modeling, offering a unique opportunity to replicate physiological tissue organization and functionality. This study investigates the impact of radiation on skeletal muscle response using an innovative in vitro human 3D skeletal muscle organoids (hSMOs) model derived from hPSCs. METHODS The hSMOs model was established through a differentiation protocol faithfully recapitulating embryonic myogenesis and maturation via paraxial mesodermal differentiation of hPSCs. Key skeletal muscle characteristics were confirmed using immunofluorescent staining and RT-qPCR. Subsequently, the hSMOs were exposed to a clinically relevant dose of 2 Gy of radiation, and their response was analyzed using immunofluorescent staining and RNA-seq. RESULTS The hSMO model faithfully recapitulated embryonic myogenesis and maturation, maintaining key skeletal muscle characteristics. Following exposure to 2 Gy of radiation, histopathological analysis revealed deficits in hSMOs expansion, differentiation, and repair response across various cell types at early (30 min) and intermediate (18 h) time points post-radiation. Immunofluorescent staining targeting γH2AX and 53BP1 demonstrated elevated levels of foci per cell, particularly in PAX7+ cells, during early and intermediate time points, with a distinct kinetic pattern showing a decrease at 72 h. RNA-seq data provided comprehensive insights into the DNA damage response within the hSMOs. CONCLUSIONS Our findings highlight deficits in expansion, differentiation, and repair response in hSMOs following radiation exposure, enhancing our understanding of radiation effects on skeletal muscle and contributing to strategies for mitigating radiation-induced damage in this context.
Collapse
Affiliation(s)
- Yifei Jiang
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Runtao Zhou
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Fawei Liao
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Ganggang Kong
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China; Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Jingguang Zeng
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Yixun Wu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Xubo Li
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Bo Wang
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Fangze Qi
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Shiju Chen
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Qintang Zhu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Liqiang Gu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Canbin Zheng
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China.
| |
Collapse
|
2
|
Park SWS, Fransson S, Sundquist F, Nilsson JN, Grybäck P, Wessman S, Strömgren J, Djos A, Fagman H, Sjögren H, Georgantzi K, Herold N, Kogner P, Granberg D, Gaze MN, Martinsson T, Karlsson K, Stenman JJE. Heterogeneous SSTR2 target expression and a novel KIAA1549:: BRAF fusion clone in a progressive metastatic lesion following 177Lutetium-DOTATATE molecular radiotherapy in neuroblastoma: a case report. Front Oncol 2024; 14:1408729. [PMID: 39324010 PMCID: PMC11422106 DOI: 10.3389/fonc.2024.1408729] [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: 03/28/2024] [Accepted: 06/20/2024] [Indexed: 09/27/2024] Open
Abstract
In this case report, we present the treatment outcomes of the first patient enrolled in the LuDO-N trial. The patient is a 21-month-old girl diagnosed with high-risk neuroblastoma (NB) and widespread skeletal metastasis. The patient initially underwent first-line therapy according to SIOPEN HRNBL-1 but was switched to second-line treatments due to disease progression, and she was finally screened for enrollment in the LuDO-N trial due to refractory disease. Upon enrollment, the patient received two rounds of the radiolabeled somatostatin analogue lutetium-177 octreotate (177Lu-DOTATATE), which was well tolerated. A dosimetry analysis revealed a heterogeneous uptake across tumor lesions, resulting in a significant absorbed dose of 54 Gy in the primary tumor, but only 2 Gy at one of the metastatic sites in the distal femur. While the initial treatment response showed disease stabilization, the distal femoral metastasis continued to progress, leading to the eventual death of the patient. A tissue analysis of the biopsies collected throughout the course of the disease revealed heterogeneous drug target expression of somatostatin receptor 2 (SSTR2) across and within tumor lesions. Furthermore, genomic profiling revealed a novel KIAA1549::BRAF fusion oncogene amplification in the distal femoral metastasis at recurrence that might be related with resistance to radiation, possibly through the downregulation of SSTR2. This case report demonstrates a mixed response to molecular radiotherapy (MRT) with 177Lu-DOTATATE. The observed variation in SSTR2 expression between tumor lesions suggests that heterogeneous target expression may have been the reason for treatment failure in this patient's case. Further investigation within the LuDO-N trial will give a more comprehensive understanding of the correlation between SSTR2 expression levels and treatment outcomes, which will be important to advance treatment strategies based on MRT for children with high-risk NB.
Collapse
Affiliation(s)
- Se Whee Sammy Park
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Fransson
- Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Sundquist
- Department of Urology, Karolinska University Hospital, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Joachim N Nilsson
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Per Grybäck
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Wessman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Jacob Strömgren
- Department of Pediatric Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Djos
- Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Fagman
- Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Helene Sjögren
- Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kleopatra Georgantzi
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Nikolas Herold
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Per Kogner
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Dan Granberg
- Department of Breast, Endocrine Tumors and Sarcomas, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Mark N Gaze
- Department of Oncology, University College London Hospitals National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Tommy Martinsson
- Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kasper Karlsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Jakob J E Stenman
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatric Surgery, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
3
|
Jiang Y, Zhou R, Wu Y, Kong G, Zeng J, Li X, Wang B, Gu C, Liao F, Qi F, Zhu Q, Gu L, Zheng C. In vitro modeling of skeletal muscle ischemia-reperfusion injury based on sphere differentiation culture from human pluripotent stem cells. Exp Cell Res 2024; 439:114111. [PMID: 38823471 DOI: 10.1016/j.yexcr.2024.114111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/14/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Skeletal muscle ischemia-reperfusion (IR) injury poses significant challenges due to its local and systemic complications. Traditional studies relying on two-dimensional (2D) cell culture or animal models often fall short of faithfully replicating the human in vivo environment, thereby impeding the translational process from animal research to clinical applications. Three-dimensional (3D) constructs, such as skeletal muscle spheroids with enhanced cell-cell interactions from human pluripotent stem cells (hPSCs) offer a promising alternative by partially mimicking human physiological cellular environment in vivo processes. This study aims to establish an innovative in vitro model, human skeletal muscle spheroids based on sphere differentiation from hPSCs, to investigate human skeletal muscle developmental processes and IR mechanisms within a controlled laboratory setting. By eticulously recapitulating embryonic myogenesis through paraxial mesodermal differentiation of neuro-mesodermal progenitors, we successfully established 3D skeletal muscle spheroids that mirror the dynamic colonization observed during human skeletal muscle development. Co-culturing human skeletal muscle spheroids with spinal cord spheroids facilitated the formation of neuromuscular junctions, providing functional relevance to skeletal muscle spheroids. Furthermore, through oxygen-glucose deprivation/re-oxygenation treatment, 3D skeletal muscle spheroids provide insights into the molecular events and pathogenesis of IR injury. The findings presented in this study significantly contribute to our understanding of skeletal muscle development and offer a robust platform for in vitro studies on skeletal muscle IR injury, holding potential applications in drug testing, therapeutic development, and personalized medicine within the realm of skeletal muscle-related pathologies.
Collapse
Affiliation(s)
- Yifei Jiang
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Runtao Zhou
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Yixun Wu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Ganggang Kong
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China; Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingguang Zeng
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Xubo Li
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Bo Wang
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Cheng Gu
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China; Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fawei Liao
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Fangze Qi
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Qintang Zhu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Liqiang Gu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Canbin Zheng
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China.
| |
Collapse
|
4
|
Mohammadi M, Banisharif S, Moradi F, Zamanian M, Tanzifi G, Ghaderi S. Brain diffusion MRI biomarkers after oncology treatments. Rep Pract Oncol Radiother 2024; 28:823-834. [PMID: 38515826 PMCID: PMC10954263 DOI: 10.5603/rpor.98728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 12/04/2023] [Indexed: 03/23/2024] Open
Abstract
In addition to providing a measurement of the tumor's size and dimensions, magnetic resonance imaging (MRI) provides excellent noninvasive radiographic detection of tumor location. The MRI technique is an important modality that has been shown to be useful in the prognosis, diagnosis, treatment planning, and evaluation of response and recurrence in solid cancers. Diffusion-weighted imaging (DWI) is an imaging technique that quantifies water mobility. This imaging approach is good for identifying sub-voxel microstructure of tissues, correlates with tumor cellularity, and has been proven to be valuable in the early assessment of cytotoxic treatment for a variety of malignancies. Diffusion tensor imaging (DTI) is an MRI method that assesses the preferred amount of water transport inside tissues. This enables precise measurements of water diffusion, which changes according to the direction of white matter fibers, their density, and myelination. This measurement corresponds to some related variables: fractional anisotropy (FA), mean diffusivity (MD), radial diffusivity (RD), axial diffusivity (AD), and others. DTI biomarkers can detect subtle changes in white matter microstructure and integrity following radiation therapy (RT) or chemoradiotherapy, which may have implications for cognitive function and quality of life. In our study, these indices were evaluated after brain chemoradiotherapy.
Collapse
Affiliation(s)
- Mahdi Mohammadi
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shabnam Banisharif
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Fatemeh Moradi
- Department of Energy Engineering & Physics, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Maryam Zamanian
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Ghazal Tanzifi
- Department of Nuclear Engineering, Islamic Azad University, Central Tehran Branch, Tehran, Iran
| | - Sadegh Ghaderi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Pathak B, Lange TE, Lampe K, Hollander E, Oria M, Murphy KP, Salomonis N, Sertorio M, Oria M. Development of a Single-Neurosphere Culture to Assess Radiation Toxicity and Pre-Clinical Cancer Combination Therapy Safety. Cancers (Basel) 2023; 15:4916. [PMID: 37894283 PMCID: PMC10605382 DOI: 10.3390/cancers15204916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Radiation therapy (RT) is a crucial treatment modality for central nervous system (CNS) tumors but toxicity to healthy CNS tissues remains a challenge. Additionally, environmental exposure to radiation during nuclear catastrophes or space travel presents a risk of CNS toxicity. However, the underlying mechanisms of radiation-induced CNS toxicity are not fully understood. Neural progenitor cells (NPCs) are highly radiosensitive, resulting in decreased neurogenesis in the hippocampus. This study aimed to characterize a novel platform utilizing rat NPCs cultured as 3D neurospheres (NSps) to screen the safety and efficacy of experimental drugs with and without radiation exposure. The effect of radiation on NSp growth and differentiation was assessed by measuring sphere volume and the expression of neuronal differentiation markers Nestin and GFAP and proliferation marker Ki67. Radiation exposure inhibited NSp growth, decreased proliferation, and increased GFAP expression, indicating astrocytic differentiation. RNA sequencing analysis supported these findings, showing upregulation of Notch, BMP2/4, S100b, and GFAP gene expression during astrogenesis. By recapitulating radiation-induced toxicity and astrocytic differentiation, this single-NSp culture system provides a high-throughput preclinical model for assessing the effects of various radiation modalities and evaluates the safety and efficacy of potential therapeutic interventions in combination with radiation.
Collapse
Affiliation(s)
- Bedika Pathak
- Center for Fetal and Placental Research, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA; (B.P.); (K.L.)
| | - Taylor E. Lange
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA
| | - Kristin Lampe
- Center for Fetal and Placental Research, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA; (B.P.); (K.L.)
| | - Ella Hollander
- Center for Fetal and Placental Research, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA; (B.P.); (K.L.)
| | - Marina Oria
- Center for Fetal and Placental Research, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA; (B.P.); (K.L.)
| | - Kendall P. Murphy
- Center for Fetal and Placental Research, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA; (B.P.); (K.L.)
- Department of Orthopedic Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA;
- Departments of Pediatrics and Bioinformatics, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Mathieu Sertorio
- University of Cincinnati Cancer Center, Cincinnati, OH 45267, USA;
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Marc Oria
- University of Cincinnati Cancer Center, Cincinnati, OH 45267, USA;
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- University of Cincinnati Brain Tumor Center, Cincinnati, OH 45219, USA
| |
Collapse
|
6
|
Profiling of gene expression in the brain associated with anxiety-related behaviors in the chronic phase following cranial irradiation. Sci Rep 2022; 12:13162. [PMID: 35915120 PMCID: PMC9343641 DOI: 10.1038/s41598-022-17310-z] [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: 02/06/2022] [Accepted: 07/22/2022] [Indexed: 11/08/2022] Open
Abstract
Although the brain is exposed to cranial irradiation in many clinical contexts, including malignant brain tumor therapy, such exposure can cause delayed neuropsychiatric disorders in the chronic phase. However, how specific molecular mechanisms are associated with irradiation-induced behavioral dysfunction, especially anxiety-like behaviors, is unclear. In the present study, we evaluated anxiety-like behaviors in adult C57BL/6 mice using the open-field (OF) and elevated plus maze (EPM) tests 3 months following single cranial irradiation (10 Gy). Additionally, by using RNA sequencing (RNA-seq), we analyzed gene expression profiles in the cortex and hippocampus of the adult brain to demonstrate the molecular mechanisms of radiation-induced brain dysfunction. In the OF and EPM tests, mice treated with radiation exhibited increased anxiety-like behaviors in the chronic phase. Gene expression analysis by RNA-seq revealed 89 and 106 differentially expressed genes in the cortex and hippocampus, respectively, following cranial irradiation. Subsequently, ClueGO and STRING analyses clustered these genes in pathways related to protein kinase activity, circadian behavior, and cell differentiation. Based on our expression analysis, we suggest that behavioral dysfunction following cranial irradiation is associated with altered expression of Cdkn1a, Ciart, Fos, Hspa5, Hspb1 and Klf10. These novel findings may provide potential genetic targets to investigate for the development of radioprotective agents.
Collapse
|
7
|
Mercer-Smith AR, Buckley A, Valdivia A, Jiang W, Thang M, Bell N, Kumar RJ, Bomba HN, Woodell AS, Luo J, Floyd SR, Hingtgen SD. Next-generation Tumor-homing Induced Neural Stem Cells as an Adjuvant to Radiation for the Treatment of Metastatic Lung Cancer. Stem Cell Rev Rep 2022; 18:2474-2493. [PMID: 35441348 DOI: 10.1007/s12015-022-10375-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2022] [Indexed: 10/18/2022]
Abstract
The spread of non-small cell lung cancer (NSCLC) to the leptomeninges is devastating with a median survival of only a few months. Radiation offers symptomatic relief, but new adjuvant therapies are desperately needed. Spheroidal, human induced neural stem cells (hiNeuroS) secreting the cytotoxic protein, TRAIL, have innate tumoritropic properties. Herein, we provide evidence that hiNeuroS-TRAIL cells can migrate to and suppress growth of NSCLC metastases in combination with radiation. In vitro cell tracking and post-mortem tissue analysis showed that hiNeuroS-TRAIL cells migrate to NSCLC tumors. Importantly, isobolographic analysis suggests that TRAIL with radiation has a synergistic cytotoxic effect on NSCLC tumors. In vivo, mice treated with radiation and hiNeuroS-TRAIL showed significant (36.6%) improvements in median survival compared to controls. Finally, bulk mRNA sequencing analysis showed both NSCLC and hiNeuroS-TRAIL cells showed changes in genes involved in migration following radiation. Overall, hiNeuroS-TRAIL cells +/- radiation have the capacity to treat NSCLC metastases.
Collapse
Affiliation(s)
- Alison R Mercer-Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Andrew Buckley
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alain Valdivia
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Wulin Jiang
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Morrent Thang
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Noah Bell
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Rashmi J Kumar
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Hunter N Bomba
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alex S Woodell
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jie Luo
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Scott R Floyd
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Shawn D Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. .,Department of Neurosurgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
8
|
Rocheteau P, Warot G, Chapellier M, Zampaolo M, Chretien F, Piquemal F. Cryopreserved Stem Cells Incur Damages Due To Terrestrial Cosmic Rays Impairing Their Integrity Upon Long-Term Storage. Cell Transplant 2022; 31:9636897211070239. [PMID: 35170351 PMCID: PMC8855380 DOI: 10.1177/09636897211070239] [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] [Indexed: 12/05/2022] Open
Abstract
Stem cells have the capacity to ensure the renewal of tissues and organs. They
could be used in the future for a wide range of therapeutic purposes and are
preserved at liquid nitrogen temperature to prevent any chemical or biological
activity up to several decades before their use. We show that the cryogenized
cells accumulate damages coming from natural radiations, potentially inducing
DNA double-strand breaks (DSBs). Such DNA damage in stem cells could lead to
either mortality of the cells upon thawing or a mutation diminishing the
therapeutic potential of the treatment. Many studies show how stem cells react
to different levels of radiation; the effect of terrestrial cosmic rays being
key, it is thus also important to investigate the effect of the natural
radiation on the cryopreserved stem cell behavior over time. Our study showed
that the cryostored stem cells totally shielded from cosmic rays had less DSBs
upon long-term storage. This could have important implications on the long-term
cryostorage strategy and quality control of different cell banks.
Collapse
Affiliation(s)
- P Rocheteau
- Human Histopathology and Animal Models, Department of Infection & Epidemiology, Institut Pasteur, Paris, France
| | - G Warot
- Laboratoire de Physique Subatomique et Corpusculaire, UMR 5821, Université Grenoble Alpes, Centre National de la Recherche Scientifique, Grenoble Institute of Technology (Institute of Engineering University Grenoble Alpes), LPSC-IN2P3, Grenoble, France
| | - M Chapellier
- Laboratoire de Physique Subatomique et Corpusculaire, UMR 5821, Université Grenoble Alpes, Centre National de la Recherche Scientifique, Grenoble Institute of Technology (Institute of Engineering University Grenoble Alpes), LPSC-IN2P3, Grenoble, France
| | - M Zampaolo
- Laboratoire de Physique Subatomique et Corpusculaire, UMR 5821, Université Grenoble Alpes, Centre National de la Recherche Scientifique, Grenoble Institute of Technology (Institute of Engineering University Grenoble Alpes), LPSC-IN2P3, Grenoble, France
| | - F Chretien
- Human Histopathology and Animal Models, Department of Infection & Epidemiology, Institut Pasteur, Paris, France
| | - F Piquemal
- Centre d'Etudes Nucléaires de Bordeaux Gradignan, UMR 5797, Centre National de la Recherche Scientifique and Université de Bordeaux, Gradignan, France
| |
Collapse
|
9
|
Ayoub R, Lau K, Yuen N, Fernandes D, Elder M, Yeung J, Wong SC, Nieman BJ. Spatiotemporal Mapping of Early Volume Loss in the Mouse Brain after Cranial Irradiation. Radiat Res 2021; 196:394-403. [PMID: 34270782 DOI: 10.1667/rade-21-00013.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 07/05/2021] [Indexed: 11/03/2022]
Abstract
Sequelae after pediatric cranial radiotherapy (CRT) result in long-term changes in brain structure. While past evidence indicates regional differences in brain volume change, it remains unclear how these manifest in the time course of change after CRT. In this study, we spatiotemporally characterized volume losses induced by cranial irradiation in a mouse model, with a dense sampling of measurements over the first week postirradiation. Wild-type mice received whole-brain irradiation (7 Gy) or sham irradiation (0 Gy) at 16 days of age. In vivo magnetic resonance imaging was performed at one time point before, and 2-4 time points postirradiation in each mouse, with a particular focus on sampling during the first week after cranial irradiation. Volume changes across the brain were measured, and the degree and timing of volume loss were quantified across structures from a predefined atlas. Volume measurements across the brain after cranial irradiation revealed a ∼2-day delay in which volume is not significantly altered, after which time volume change proceeds over the course of four days. Volume losses were 3% larger and emerged 40% slower in white matter than in gray matter. Large volume loss was also observed in the ventricles. Differences in the timing and magnitude of volume change between gray and white matter after cranial irradiation were observed. These results suggest differences in the mechanism and/or kinetics underlying the associated radio-response, which may have implications in development.
Collapse
Affiliation(s)
- Ramy Ayoub
- Departments of Medical Biophysics, University of Toronto, Ontario, Canada
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kaylie Lau
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nili Yuen
- Departments of Medical Biophysics, University of Toronto, Ontario, Canada
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Darren Fernandes
- Departments of Medical Biophysics, University of Toronto, Ontario, Canada
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Madeline Elder
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jonas Yeung
- Departments of Medical Biophysics, University of Toronto, Ontario, Canada
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Shun C Wong
- Departments of Medical Biophysics, University of Toronto, Ontario, Canada
- Departments of Radiation Oncology, University of Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Ontario, Canada
- The Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Brian J Nieman
- Departments of Medical Biophysics, University of Toronto, Ontario, Canada
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Bhat K, Medina P, He L, Zhang L, Saki M, Ioannidis A, Nguyen NT, Sodhi SS, Sung D, Magyar CE, Liau LM, Kornblum HI, Pajonk F. 1-[(4-Nitrophenyl)sulfonyl]-4-phenylpiperazine treatment after brain irradiation preserves cognitive function in mice. Neuro Oncol 2021; 22:1484-1494. [PMID: 32291451 DOI: 10.1093/neuonc/noaa095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Normal tissue toxicity is an inevitable consequence of primary or secondary brain tumor radiotherapy. Cranial irradiation commonly leads to neurocognitive deficits that manifest months or years after treatment. Mechanistically, radiation-induced loss of neural stem/progenitor cells, neuroinflammation, and demyelination are contributing factors that lead to progressive cognitive decline. METHODS The effects of 1-[(4-nitrophenyl)sulfonyl]-4-phenylpiperazine (NSPP) on irradiated murine neurospheres, microglia cells, and patient-derived gliomaspheres were assessed by sphere-formation assays, flow cytometry, and interleukin (IL)-6 enzyme-linked immunosorbent assay. Activation of the hedgehog pathway was studied by quantitative reverse transcription PCR. The in vivo effects of NSPP were analyzed using flow cytometry, sphere-formation assays, immunohistochemistry, behavioral testing, and an intracranial mouse model of glioblastoma. RESULTS We report that NSPP mitigates radiation-induced normal tissue toxicity in the brains of mice. NSPP treatment significantly increased the number of neural stem/progenitor cells after brain irradiation in female animals, and inhibited radiation-induced microglia activation and expression of the pro-inflammatory cytokine IL-6. Behavioral testing revealed that treatment with NSPP after radiotherapy was able to successfully mitigate radiation-induced decline in memory function of the brain. In mouse models of glioblastoma, NSPP showed no toxicity and did not interfere with the growth-delaying effects of radiation. CONCLUSIONS We conclude that NSPP has the potential to mitigate cognitive decline in patients undergoing partial or whole brain irradiation without promoting tumor growth and that the use of this compound as a radiation mitigator of radiation late effects on the central nervous system warrants further investigation.
Collapse
Affiliation(s)
- Kruttika Bhat
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Paul Medina
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Ling He
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Le Zhang
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Mohammad Saki
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Angeliki Ioannidis
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Nhan T Nguyen
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Sirajbir S Sodhi
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - David Sung
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Clara E Magyar
- Translational Pathology Core Laboratory, Image Analysis/Virtual Microscopy, Department of Pathology and Laboratory Medicine, Los Angeles, California
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California.,Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
| | - Harley I Kornblum
- NPI-Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, California.,Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California.,Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
| |
Collapse
|
11
|
New Discoveries in Radiation Science. Cancers (Basel) 2021; 13:cancers13051034. [PMID: 33801176 PMCID: PMC7957593 DOI: 10.3390/cancers13051034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/18/2021] [Accepted: 02/23/2021] [Indexed: 01/15/2023] Open
|
12
|
Coco S, Boccardo S, Mora M, Fontana V, Vanni I, Genova C, Alama A, Salvi S, Dal Bello MG, Bonfiglio S, Rijavec E, Sini C, Barletta G, Biello F, Carli F, Cavalieri Z, Burrafato G, Longo L, Ballestrero A, Grossi F. Radiation-Related Deregulation of TUBB3 and BRCA1/2 and Risk of Secondary Lung Cancer in Women With Breast Cancer. Clin Breast Cancer 2020; 21:218-230.e6. [PMID: 33008754 DOI: 10.1016/j.clbc.2020.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/05/2020] [Accepted: 09/02/2020] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Breast cancer survivors are at increased risk of developing unrelated primary cancers, particularly lung cancer. Evidence indicates that sex hormones as well as a deregulation of DNA-repair pathways may contribute to lung cancer onset. We investigated whether the hormone status and expression of markers involved in DNA repair (BRCA1/2, ERCC1, and P53R2), synthesis (TS and RRM1), and cell division (TUBB3) might be linked to lung cancer risk. PATIENTS AND METHODS Thirty-seven breast cancer survivors with unrelated lung cancer and 84 control subjects comprising women with breast cancer (42/84) or lung cancer (42/84) were enrolled. Immunohistochemistry on tumor tissue was performed. Geometric mean ratio was used to assess the association of marker levels with patient groups. RESULTS Estrogen receptor was expressed in approximately 90% of the breast cancer group but was negative in the majority of the lung cancer group, a result similar to the lung cancer control group. Likewise, ER isoform β was weakly expressed in the lung cancer group. Protein analysis of breast cancer versus control had a significantly lower expression of BRCA1, P53R2, and TUBB3. Likewise, a BRCA1 reduction was observed in the lung cancer group concomitant with a BRCA2 increase. Furthermore, BRCA2 and TUBB3 increased in ipsilateral lung cancer in women who had previously received radiotherapy for breast cancer. CONCLUSION The decrease of DNA-repair proteins in breast cancer could make these women more susceptible to therapy-related cancer. The increase of BRCA2 and TUBB3 in lung cancer from patients who previously received radiotherapy for breast cancer might reflect a tissue response to exposure to ionizing radiation.
Collapse
Affiliation(s)
- Simona Coco
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Simona Boccardo
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Vincenzo Fontana
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Irene Vanni
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa
| | - Carlo Genova
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa
| | - Angela Alama
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | | | - Silvia Bonfiglio
- Centre for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Erika Rijavec
- UOC Oncologia Medica, IRCCS Cà Granda Foundation, Ospedale Maggiore Policlinico, Milan, Italy
| | - Claudio Sini
- Oncologia Medica e CPDO, ASSL di Olbia-ATS Sardegna, Olbia, Italy
| | - Giulia Barletta
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | | | - Zita Cavalieri
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Luca Longo
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Alberto Ballestrero
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa
| | - Francesco Grossi
- UOC Oncologia Medica, IRCCS Cà Granda Foundation, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
13
|
X-ray irradiated cultures of mouse cortical neural stem/progenitor cells recover cell viability and proliferation with dose-dependent kinetics. Sci Rep 2020; 10:6562. [PMID: 32300147 PMCID: PMC7162981 DOI: 10.1038/s41598-020-63348-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 03/24/2020] [Indexed: 12/13/2022] Open
Abstract
Exposure of the developing or adult brain to ionizing radiation (IR) can cause cognitive impairment and/or brain cancer, by targeting neural stem/progenitor cells (NSPCs). IR effects on NSPCs include transient cell cycle arrest, permanent cell cycle exit/differentiation, or cell death, depending on the experimental conditions. In vivo studies suggest that brain age influences NSPC response to IR, but whether this is due to intrinsic NSPC changes or to niche environment modifications remains unclear. Here, we describe the dose-dependent, time-dependent effects of X-ray IR in NSPC cultures derived from the mouse foetal cerebral cortex. We show that, although cortical NSPCs are resistant to low/moderate IR doses, high level IR exposure causes cell death, accumulation of DNA double-strand breaks, activation of p53-related molecular pathways and cell cycle alterations. Irradiated NSPC cultures transiently upregulate differentiation markers, but recover control levels of proliferation, viability and gene expression in the second week post-irradiation. These results are consistent with previously described in vivo effects of IR in the developing mouse cortex, and distinct from those observed in adult NSPC niches or in vitro adult NSPC cultures, suggesting that intrinsic differences in NSPCs of different origins might determine, at least in part, their response to IR.
Collapse
|
14
|
Wang Z, Zheng Y, Zheng M, Zhong J, Ma F, Zhou B, Zhu J. Neurogenic Niche Conversion Strategy Induces Migration and Functional Neuronal Differentiation of Neural Precursor Cells Following Brain Injury. Stem Cells Dev 2020; 29:235-248. [PMID: 31797735 DOI: 10.1089/scd.2019.0147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glial scars formed after brain injuries provide permissive cues for endogenous neural precursor/stem cells (eNP/SCs) to undergo astrogenesis rather than neurogenesis. Following brain injury, eNP/SCs from the subventricular zone leave their niche, migrate to the injured cortex, and differentiate into reactive astrocytes that contribute to glial scar formation. In vivo neuronal reprogramming, directly converting non-neuronal cells such as reactive astrocytes or NG2 glia into neurons, has greatly improved brain injury repair strategies. However, reprogramming carries a high risk of future clinical applications such as tumorigenicity, involving virus. In this study, we constructed a neural matrix to alter the adverse niche at the injured cortex, enabling eNP/SCs to differentiate into functional neurons. We found that the neural matrix functioned as a "glial trap" that largely concentrated and limited reactive astrocytes to the core of the lesion area, thus altering the adverse niche. The eNP/SCs migrated toward the injured cortex and differentiated into functional neurons. In addition, regenerated neurites extended across the boundary of the injured cortex. Mice treated with the neural matrix demonstrated significant behavioral recovery. For the first time, we induced eNP/SC-derived functional neurons in the cortex after brain injury without the use of viruses, microRNAs, or small molecules. Our novel strategy of applying this "glial trap" to obtain functional neurons in the injured cortex may provide a safer and more natural therapeutic alternative to reprogramming in future clinical applications.
Collapse
Affiliation(s)
- Zhifu Wang
- Department of Neurosurgery, Huashan Hospital and National Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yongtao Zheng
- Department of Neurosurgery, Huashan Hospital and National Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mingzhe Zheng
- Department of Neurosurgery, Huashan Hospital and National Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Junjie Zhong
- Department of Neurosurgery, Huashan Hospital and National Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fukai Ma
- Department of Neurosurgery, Huashan Hospital and National Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgery Department, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (CAS), University of Chinese Academy of Sciences, Shanghai, China
| | - Jianhong Zhu
- Department of Neurosurgery, Huashan Hospital and National Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Michaelidesová A, Konířová J, Bartůněk P, Zíková M. Effects of Radiation Therapy on Neural Stem Cells. Genes (Basel) 2019; 10:E640. [PMID: 31450566 PMCID: PMC6770913 DOI: 10.3390/genes10090640] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/19/2019] [Accepted: 08/22/2019] [Indexed: 12/29/2022] Open
Abstract
Brain and nervous system cancers in children represent the second most common neoplasia after leukemia. Radiotherapy plays a significant role in cancer treatment; however, the use of such therapy is not without devastating side effects. The impact of radiation-induced damage to the brain is multifactorial, but the damage to neural stem cell populations seems to play a key role. The brain contains pools of regenerative neural stem cells that reside in specialized neurogenic niches and can generate new neurons. In this review, we describe the advances in radiotherapy techniques that protect neural stem cell compartments, and subsequently limit and prevent the occurrence and development of side effects. We also summarize the current knowledge about neural stem cells and the molecular mechanisms underlying changes in neural stem cell niches after brain radiotherapy. Strategies used to minimize radiation-related damages, as well as new challenges in the treatment of brain tumors are also discussed.
Collapse
Affiliation(s)
- Anna Michaelidesová
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the Czech Academy of Sciences, v. v. i., Vídeňská 1083, 142 20 Prague 4, Czech Republic
- Department of Radiation Dosimentry, Nuclear Physics Institute of the Czech Academy of Sciences, v. v. i., Na Truhlářce 39/64, 180 00 Prague 8, Czech Republic
| | - Jana Konířová
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the Czech Academy of Sciences, v. v. i., Vídeňská 1083, 142 20 Prague 4, Czech Republic
- Department of Radiation Dosimentry, Nuclear Physics Institute of the Czech Academy of Sciences, v. v. i., Na Truhlářce 39/64, 180 00 Prague 8, Czech Republic
| | - Petr Bartůněk
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the Czech Academy of Sciences, v. v. i., Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Martina Zíková
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the Czech Academy of Sciences, v. v. i., Vídeňská 1083, 142 20 Prague 4, Czech Republic.
| |
Collapse
|