1
|
Li J, Sun B, Tan LX, Griffin N, Niknezhad SV, Yu C, Berthoin L, Cruz-Pacheco N, Mohabbat S, Sinada H, Efraim Y, Chen FYT, An L, Gaylord EA, Bahney CS, Lombaert IM, Knox SM. Rescue of non-healing, degenerative salivary glands by cholinergic-calcium signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.31.630834. [PMID: 39803569 PMCID: PMC11722244 DOI: 10.1101/2024.12.31.630834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Chronic degenerative wounds are often deemed irreparable, directing research efforts to focus predominantly on acute tissue injury regeneration while leaving endogenous repair mechanisms for chronically damaged tissues largely unexplored. In this study, we demonstrate that non-healing, severely degenerated salivary gland tissues can be fundamentally restored through first-line treatment with muscarinic agonists. This approach rescues tissue structure and function, returning it to a homeostatic-like state, and reactivates endogenous regeneration processes to drive new cell expansion that persists for months post-treatment. Furthermore, neuromimetic activation profoundly depletes radiation-induced DNA damage and re-establishes the nerve-acinar relationship, ultimately restoring the tissues physiological capacity to maintain homeostasis, even in the absence of treatment. We show that full recovery of organ function, comparable to uninjured controls, is primarily mediated by the re-differentiation of aberrantly de-differentiated epithelial acinar cells and the restoration of mitochondrial function via a muscarinic-calcium signaling pathway. These findings challenge the prevailing notion that chronic organ degeneration is irreversible and propose a readily testable therapeutic strategy for epithelial restoration with potential applications across a spectrum of chronic injuries.
Collapse
Affiliation(s)
- Jianlong Li
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
- These authors contributed equally
| | - Bo Sun
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
- These authors contributed equally
| | - Li Xuan Tan
- Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, California, USA; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
- These authors contributed equally
| | - Nathan Griffin
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Seyyed Vahid Niknezhad
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Chieh Yu
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Lionel Berthoin
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Noel Cruz-Pacheco
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Seayar Mohabbat
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Hanan Sinada
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Yael Efraim
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Feeling Yu Ting Chen
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Luye An
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Eliza A. Gaylord
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Chelsey S. Bahney
- University of California, San Francisco. Orthopedic Trauma Institute, San Francisco, CA
| | - Isabelle M.A. Lombaert
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA
- Co–senior authors
| | - Sarah M. Knox
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
- Co–senior authors
- Lead contact
| |
Collapse
|
2
|
Johnson AL, Elder SS, McKendrick JG, Hegarty LM, Mercer E, Emmerson E. A single dose of radiation elicits comparable acute salivary gland injury to fractionated radiation. Dis Model Mech 2024; 17:dmm050733. [PMID: 39086326 PMCID: PMC11361643 DOI: 10.1242/dmm.050733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
The salivary glands are often damaged during head and neck cancer radiotherapy. This results in chronic dry mouth, which adversely affects quality of life and for which there is no long-term cure. Mouse models of salivary gland injury are routinely used in regenerative research. However, there is no clear consensus on the radiation regime required to cause injury. Here, we analysed three regimes of γ-irradiation of the submandibular salivary gland. Transcriptional analysis, immunofluorescence and flow cytometry was used to profile DNA damage, gland architecture and immune cell changes 3 days after single doses of 10 or 15 Gy or three doses of 5 Gy. Irrespective of the regime, radiation induced comparable levels of DNA damage, cell cycle arrest, loss of glandular architecture, increased pro-inflammatory cytokines and a reduction in tissue-resident macrophages, relative to those observed in non-irradiated submandibular glands. Given these data, coupled with the fact that repeated anaesthetic can negatively affect animal welfare and interfere with saliva secretion, we conclude that a single dose of 10 Gy irradiation is the most refined method of inducing acute salivary gland injury in a mouse model.
Collapse
Affiliation(s)
- Amanda L. Johnson
- The Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Sonia S. Elder
- The Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - John G. McKendrick
- The Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
- The Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Lizi M. Hegarty
- The Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
- The Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Ella Mercer
- The Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Elaine Emmerson
- The Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| |
Collapse
|
3
|
Gunning JA, Gilman KE, Zúñiga TM, Simpson RJ, Limesand KH. Parotid glands have a dysregulated immune response following radiation therapy. PLoS One 2024; 19:e0297387. [PMID: 38470874 PMCID: PMC10931461 DOI: 10.1371/journal.pone.0297387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 01/04/2024] [Indexed: 03/14/2024] Open
Abstract
Head and neck cancer treatment often consists of surgical resection of the tumor followed by ionizing radiation (IR), which can damage surrounding tissues and cause adverse side effects. The underlying mechanisms of radiation-induced salivary gland dysfunction are not fully understood, and treatment options are scarce and ineffective. The wound healing process is a necessary response to tissue injury, and broadly consists of inflammatory, proliferative, and redifferentiation phases with immune cells playing key roles in all three phases. In this study, select immune cells were phenotyped and quantified, and certain cytokine and chemokine concentrations were measured in mouse parotid glands after IR. Further, we used a model where glandular function is restored to assess the immune phenotype in a regenerative response. These data suggest that irradiated parotid tissue does not progress through a typical inflammatory response observed in wounds that heal. Specifically, total immune cells (CD45+) decrease at days 2 and 5 following IR, macrophages (F4/80+CD11b+) decrease at day 2 and 5 and increase at day 30, while neutrophils (Ly6G+CD11b+) significantly increase at day 30 following IR. Additionally, radiation treatment reduces CD3- cells at all time points, significantly increases CD3+/CD4+CD8+ double positive cells, and significantly reduces CD3+/CD4-CD8- double negative cells at day 30 after IR. Previous data indicate that post-IR treatment with IGF-1 restores salivary gland function at day 30, and IGF-1 injections attenuate the increase in macrophages, neutrophils, and CD4+CD8+ T cells observed at day 30 following IR. Taken together, these data indicate that parotid salivary tissue exhibits a dysregulated immune response following radiation treatment which may contribute to chronic loss of function phenotype in head and neck cancer survivors.
Collapse
Affiliation(s)
- Jordan A. Gunning
- Department of Nutritional Sciences, The University of Arizona, Tucson, Arizona, United States of America
| | - Kristy E. Gilman
- Department of Nutritional Sciences, The University of Arizona, Tucson, Arizona, United States of America
| | - Tiffany M. Zúñiga
- Department of Nutritional Sciences, The University of Arizona, Tucson, Arizona, United States of America
- Department of Immunology and Microbiology, Aurora, Colorado, United States of America
| | - Richard J. Simpson
- Department of Nutritional Sciences, The University of Arizona, Tucson, Arizona, United States of America
| | - Kirsten H. Limesand
- Department of Nutritional Sciences, The University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
4
|
Buss LG, De Oliveira Pessoa D, Snider JM, Padi M, Martinez JA, Limesand KH. Metabolomics analysis of pathways underlying radiation-induced salivary gland dysfunction stages. PLoS One 2023; 18:e0294355. [PMID: 37983277 PMCID: PMC10659204 DOI: 10.1371/journal.pone.0294355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/30/2023] [Indexed: 11/22/2023] Open
Abstract
Salivary gland hypofunction is an adverse side effect associated with radiotherapy for head and neck cancer patients. This study delineated metabolic changes at acute, intermediate, and chronic radiation damage response stages in mouse salivary glands following a single 5 Gy dose. Ultra-high performance liquid chromatography-mass spectrometry was performed on parotid salivary gland tissue collected at 3, 14, and 30 days following radiation (IR). Pathway enrichment analysis, network analysis based on metabolite structural similarity, and network analysis based on metabolite abundance correlations were used to incorporate both metabolite levels and structural annotation. The greatest number of enriched pathways are observed at 3 days and the lowest at 30 days following radiation. Amino acid metabolism pathways, glutathione metabolism, and central carbon metabolism in cancer are enriched at all radiation time points across different analytical methods. This study suggests that glutathione and central carbon metabolism in cancer may be important pathways in the unresolved effect of radiation treatment.
Collapse
Affiliation(s)
- Lauren G. Buss
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, United States of America
| | - Diogo De Oliveira Pessoa
- Biostatistics and Bioinformatics Shared Resource, Arizona Cancer Center, University of Arizona, Tucson, AZ, United States of America
| | - Justin M. Snider
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, United States of America
- University of Arizona Cancer Center, Tucson, AZ, United States of America
| | - Megha Padi
- Biostatistics and Bioinformatics Shared Resource, Arizona Cancer Center, University of Arizona, Tucson, AZ, United States of America
- University of Arizona Cancer Center, Tucson, AZ, United States of America
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, United States of America
| | - Jessica A. Martinez
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, United States of America
- University of Arizona Cancer Center, Tucson, AZ, United States of America
| | - Kirsten H. Limesand
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, United States of America
- University of Arizona Cancer Center, Tucson, AZ, United States of America
| |
Collapse
|
5
|
Nieri CA, Benaim EH, Zhang YH, Garcia-Godoy F, Herr MJ, Zhang W, Schwartz D, Coca KK, Gleysteen JP, Gillespie MB. Botox for the prevention of radiation-induced Sialadenitis and xerostomia in head and neck cancer patients: A pilot study. Head Neck 2023; 45:2198-2206. [PMID: 37403447 DOI: 10.1002/hed.27449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND To determine the safety of Botox and its potential effect on alleviating radiation therapy (RT)-induced sialadenitis in head and neck cancer patients. METHODS Twenty patients with stage III/IV head and neck cancer were randomized to receive Botox or saline injections into both submandibular glands (SMG). There were three visits: one before RT (V1); 1 week after RT (V2); and 6 weeks after RT (V3), each of which included saliva collection, a 24-h dietary recall, and a quality-of-life survey. RESULTS No adverse events were observed. While the control group was much older, the Botox group more commonly underwent induction chemotherapy compared with controls. From V1 to V2, salivary flow decreased in both groups, but only in the control group from V1 to V3. CXCL-1 (GRO), a neutrophil chemoattractant, was lower in the Botox group compared with the control group at V3. CONCLUSION Botox can be safely administered to the salivary glands prior to external beam radiation without observed complications or side-effects. After an initial reduction in salivary flow following RT, the Botox group showed lack of further flow reduction compared with controls. The inflammatory marker CXCL 1, which was reduced in the in Botox group at V3, may be a candidate for further studies of radiation-induced sialadenitis.
Collapse
Affiliation(s)
- Chad Alexander Nieri
- Department of Otolaryngology- Head and Neck Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ezer Haim Benaim
- Department of Otolaryngology- Head and Neck Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Yanhui H Zhang
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Franklin Garcia-Godoy
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- The Forsyth Institute, Cambridge, Massachusetts, USA
- Department of Surgery, Herbert Wertheim, College of Medicine, Florida International University, Miami, Florida, USA
| | - Michael J Herr
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| | - Weiqiang Zhang
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - David Schwartz
- Department of Radiation Oncology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Kimberly K Coca
- Department of Otolaryngology, Head and Neck Surgery, University of South Florida, Tampa, Florida, USA
| | - John P Gleysteen
- Department of Otolaryngology- Head and Neck Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Marion Boyd Gillespie
- Department of Otolaryngology- Head and Neck Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
6
|
Zhang C, Xiang B. The underlying mechanisms and strategies of DNA damage and repair in radiation sialadenitis. Oral Dis 2023; 29:990-995. [PMID: 34773326 DOI: 10.1111/odi.14078] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/18/2021] [Accepted: 11/03/2021] [Indexed: 11/24/2022]
Abstract
Radiation therapy is a critical strategy for the treatment of malignant tumors. X-ray external radiation has been successfully used to treat head and neck cancer. On the other hand, 131 I internal radiation has been effective in managing differentiated thyroid cancer. However, these therapies cause radiation damage to salivary glands. Radiation sialadenitis is the most common complication associated with radiotherapy applied to the head and neck and it severely affects patients' quality of life. Since DNA is the main intracellular target of radiation, and the integrity of the DNA structure is critical to genomic stability and the cellular survival of salivary glands, regulating radiation-induced DNA damage offers great promise in preventing and managing radiation sialadenitis. In this review, we summarize recent progress in DNA damage and repair in irradiated salivary glands.
Collapse
Affiliation(s)
- Chong Zhang
- Laboratory of Oral and Maxillofacial Disease, The Second Hospital of Dalian Medical University, Dalian, China
| | - Bin Xiang
- Laboratory of Oral and Maxillofacial Disease, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
7
|
Juvkam IS, Zlygosteva O, Arous D, Galtung HK, Malinen E, Søland TM, Edin NJ. A preclinical model to investigate normal tissue damage following fractionated radiotherapy to the head and neck. JOURNAL OF RADIATION RESEARCH 2023; 64:44-52. [PMID: 36253091 PMCID: PMC9855321 DOI: 10.1093/jrr/rrac066] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/09/2022] [Indexed: 06/16/2023]
Abstract
Radiotherapy (RT) of head and neck (H&N) cancer is known to cause both early- and late-occurring toxicities. To better appraise normal tissue responses and their dependence on treatment parameters such as radiation field and type, as well as dose and fractionation scheme, a preclinical model with relevant endpoints is required. 12-week old female C57BL/6 J mice were irradiated with 100 or 180 kV X-rays to total doses ranging from 30 to 85 Gy, given in 10 fractions over 5 days. The radiation field covered the oral cavity, swallowing structures and salivary glands. Monte Carlo simulations were employed to estimate tissue dose distribution. The follow-up period was 35 days, in order to study the early radiation-induced effects. Baseline and post irradiation investigations included macroscopic and microscopic examinations of the skin, lips, salivary glands and oral mucosa. Saliva sampling was performed to assess the salivary gland function following radiation exposure. A dose dependent radiation dermatitis in the skin was observed for doses above 30 Gy. Oral mucositis in the tongue appeared as ulcerations on the ventral surface of the tongue for doses of 75-85 Gy. The irradiated mice showed significantly reduced saliva production compared to controls. In summary, a preclinical model to investigate a broad panel of normal tissue responses following fractionated irradiation of the H&N region was established. The optimal dose to study early radiation-induced effects was found to be around 75 Gy, as this was the highest tolerated dose that gave acute effects similar to that observed in cancer patients.
Collapse
Affiliation(s)
- Inga Solgård Juvkam
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, 0372 Oslo, Norway
| | - Olga Zlygosteva
- Department of Physics, Faculty of Mathematics and Natural Sciences, University of Oslo, 0371 Oslo, Norway
| | - Delmon Arous
- Department of Physics, Faculty of Mathematics and Natural Sciences, University of Oslo, 0371 Oslo, Norway
- Department of Medical Physics, Cancer Clinic, Oslo University Hospital, 0379 Oslo, Norway
| | - Hilde Kanli Galtung
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, 0372 Oslo, Norway
| | - Eirik Malinen
- Department of Physics, Faculty of Mathematics and Natural Sciences, University of Oslo, 0371 Oslo, Norway
- Department of Medical Physics, Cancer Clinic, Oslo University Hospital, 0379 Oslo, Norway
| | - Tine Merete Søland
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, 0372 Oslo, Norway
- Department of Pathology, Oslo University Hospital, 0372 Oslo, Norway
| | - Nina Jeppesen Edin
- Department of Physics, Faculty of Mathematics and Natural Sciences, University of Oslo, 0371 Oslo, Norway
| |
Collapse
|
8
|
Parasympathetic-macrophages-ductal epithelial cells axis promotes female rat submandibular gland regeneration after excretory duct ligation/deligation. Arch Oral Biol 2022; 145:105586. [DOI: 10.1016/j.archoralbio.2022.105586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 11/06/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
|
9
|
Chibly AM, Aure MH, Patel VN, Hoffman MP. Salivary gland function, development, and regeneration. Physiol Rev 2022; 102:1495-1552. [PMID: 35343828 PMCID: PMC9126227 DOI: 10.1152/physrev.00015.2021] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/27/2021] [Accepted: 03/17/2022] [Indexed: 02/08/2023] Open
Abstract
Salivary glands produce and secrete saliva, which is essential for maintaining oral health and overall health. Understanding both the unique structure and physiological function of salivary glands, as well as how they are affected by disease and injury, will direct the development of therapy to repair and regenerate them. Significant recent advances, particularly in the OMICS field, increase our understanding of how salivary glands develop at the cellular, molecular, and genetic levels: the signaling pathways involved, the dynamics of progenitor cell lineages in development, homeostasis, and regeneration, and the role of the extracellular matrix microenvironment. These provide a template for cell and gene therapies as well as bioengineering approaches to repair or regenerate salivary function.
Collapse
Affiliation(s)
- Alejandro M Chibly
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Marit H Aure
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Vaishali N Patel
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
10
|
Chen G, Han Y, Zhang H, Tu W, Zhang S. Radiotherapy-Induced Digestive Injury: Diagnosis, Treatment and Mechanisms. Front Oncol 2021; 11:757973. [PMID: 34804953 PMCID: PMC8604098 DOI: 10.3389/fonc.2021.757973] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Radiotherapy is one of the main therapeutic methods for treating cancer. The digestive system consists of the gastrointestinal tract and the accessory organs of digestion (the tongue, salivary glands, pancreas, liver and gallbladder). The digestive system is easily impaired during radiotherapy, especially in thoracic and abdominal radiotherapy. In this review, we introduce the physical classification, basic pathogenesis, clinical characteristics, predictive/diagnostic factors, and possible treatment targets of radiotherapy-induced digestive injury. Radiotherapy-induced digestive injury complies with the dose-volume effect and has a radiation-based organ correlation. Computed tomography (CT), MRI (magnetic resonance imaging), ultrasound (US) and endoscopy can help diagnose and evaluate the radiation-induced lesion level. The latest treatment approaches include improvement in radiotherapy (such as shielding, hydrogel spacers and dose distribution), stem cell transplantation and drug administration. Gut microbiota modulation may become a novel approach to relieving radiogenic gastrointestinal syndrome. Finally, we summarized the possible mechanisms involved in treatment, but they remain varied. Radionuclide-labeled targeting molecules (RLTMs) are promising for more precise radiotherapy. These advances contribute to our understanding of the assessment and treatment of radiation-induced digestive injury.
Collapse
Affiliation(s)
- Guangxia Chen
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Yi Han
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Haihan Zhang
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Wenling Tu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Shuyu Zhang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China.,West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Liu Z, Dong L, Zheng Z, Liu S, Gong S, Meng L, Xin Y, Jiang X. Mechanism, Prevention, and Treatment of Radiation-Induced Salivary Gland Injury Related to Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10111666. [PMID: 34829539 PMCID: PMC8614677 DOI: 10.3390/antiox10111666] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Radiation therapy is a common treatment for head and neck cancers. However, because of the presence of nerve structures (brain stem, spinal cord, and brachial plexus), salivary glands (SGs), mucous membranes, and swallowing muscles in the head and neck regions, radiotherapy inevitably causes damage to these normal tissues. Among them, SG injury is a serious adverse event, and its clinical manifestations include changes in taste, difficulty chewing and swallowing, oral infections, and dental caries. These clinical symptoms seriously reduce a patient’s quality of life. Therefore, it is important to clarify the mechanism of SG injury caused by radiotherapy. Although the mechanism of radiation-induced SG injury has not yet been determined, recent studies have shown that the mechanisms of calcium signaling, microvascular injury, cellular senescence, and apoptosis are closely related to oxidative stress. In this article, we review the mechanism by which radiotherapy causes oxidative stress and damages the SGs. In addition, we discuss effective methods to prevent and treat radiation-induced SG damage.
Collapse
Affiliation(s)
- Zijing Liu
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (Z.L.); (L.D.); (Z.Z.); (S.L.); (S.G.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Lihua Dong
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (Z.L.); (L.D.); (Z.Z.); (S.L.); (S.G.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Zhuangzhuang Zheng
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (Z.L.); (L.D.); (Z.Z.); (S.L.); (S.G.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Shiyu Liu
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (Z.L.); (L.D.); (Z.Z.); (S.L.); (S.G.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Shouliang Gong
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (Z.L.); (L.D.); (Z.Z.); (S.L.); (S.G.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China;
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (Z.L.); (L.D.); (Z.Z.); (S.L.); (S.G.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
- Correspondence: ; Tel.: +86-158-0430-2750
| |
Collapse
|
12
|
Nardone V, Barbarino M, Angrisani A, Correale P, Pastina P, Cappabianca S, Reginelli A, Mutti L, Miracco C, Giannicola R, Giordano A, Pirtoli L. CDK4, CDK6/cyclin-D1 Complex Inhibition and Radiotherapy for Cancer Control: A Role for Autophagy. Int J Mol Sci 2021; 22:8391. [PMID: 34445095 PMCID: PMC8395054 DOI: 10.3390/ijms22168391] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
The expanding clinical application of CDK4- and CDK6-inhibiting drugs in the managements of breast cancer has raised a great interest in testing these drugs in other neoplasms. The potential of combining these drugs with other therapeutic approaches seems to be an interesting work-ground to explore. Even though a potential integration of CDK4 and CDK6 inhibitors with radiotherapy (RT) has been hypothesized, this kind of approach has not been sufficiently pursued, neither in preclinical nor in clinical studies. Similarly, the most recent discoveries focusing on autophagy, as a possible target pathway able to enhance the antitumor efficacy of CDK4 and CDK6 inhibitors is promising but needs more investigations. The aim of this review is to discuss the recent literature on the field in order to infer a rational combination strategy including cyclin-D1/CDK4-CDK6 inhibitors, RT, and/or other anticancer agents targeting G1-S phase cell cycle transition.
Collapse
Affiliation(s)
- Valerio Nardone
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (A.A.); (S.C.); (A.R.)
| | - Marcella Barbarino
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (M.B.); (A.G.)
| | - Antonio Angrisani
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (A.A.); (S.C.); (A.R.)
| | - Pierpaolo Correale
- Medical Oncology Unit, Grand Metropolitan Hospital “Bianchi-Melacrino-Morelli”, 89124 Reggio Calabria, Italy; (P.C.); (R.G.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19104, USA; (L.M.); (L.P.)
| | - Pierpaolo Pastina
- Section of Radiation Oncology, Medical School, University of Siena, 53100 Siena, Italy;
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (A.A.); (S.C.); (A.R.)
| | - Alfonso Reginelli
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (A.A.); (S.C.); (A.R.)
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19104, USA; (L.M.); (L.P.)
| | - Clelia Miracco
- Pathological Anatomy Unit, Department of Medical, Surgical and Neurological Science, University of Siena, 53100 Siena, Italy;
| | - Rocco Giannicola
- Medical Oncology Unit, Grand Metropolitan Hospital “Bianchi-Melacrino-Morelli”, 89124 Reggio Calabria, Italy; (P.C.); (R.G.)
| | - Antonio Giordano
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (M.B.); (A.G.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19104, USA; (L.M.); (L.P.)
| | - Luigi Pirtoli
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19104, USA; (L.M.); (L.P.)
| |
Collapse
|
13
|
Meeks L, De Oliveira Pessoa D, Martinez JA, Limesand KH, Padi M. Integration of metabolomics and transcriptomics reveals convergent pathways driving radiation-induced salivary gland dysfunction. Physiol Genomics 2021; 53:85-98. [PMID: 33522389 PMCID: PMC7988743 DOI: 10.1152/physiolgenomics.00127.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/04/2021] [Accepted: 01/22/2021] [Indexed: 11/22/2022] Open
Abstract
Radiation therapy for head and neck cancer causes damage to the surrounding salivary glands, resulting in salivary gland hypofunction and xerostomia. Current treatments do not provide lasting restoration of salivary gland function following radiation; therefore, a new mechanistic understanding of the radiation-induced damage response is necessary for identifying therapeutic targets. The purpose of the present study was to investigate the metabolic phenotype of radiation-induced damage in parotid salivary glands by integrating transcriptomic and metabolomic data. Integrated data were then analyzed to identify significant gene-metabolite interactions. Mice received a single 5 Gy dose of targeted head and neck radiation. Parotid tissue samples were collected 5 days following treatment for RNA sequencing and metabolomics analysis. Altered metabolites and transcripts significantly converged on a specific region in the metabolic reaction network. Both integrative pathway enrichment using rank-based statistics and network analysis highlighted significantly coordinated changes in glutathione metabolism, energy metabolism (TCA cycle and thermogenesis), peroxisomal lipid metabolism, and bile acid production with radiation. Integrated changes observed in energy metabolism suggest that radiation induces a mitochondrial dysfunction phenotype. These findings validated previous pathways involved in the radiation-damage response, such as altered energy metabolism, and identified robust signatures in salivary glands, such as reduced glutathione metabolism, that may be driving salivary gland dysfunction.
Collapse
Affiliation(s)
- Lauren Meeks
- Department of Nutritional Sciences, University of Arizona, Tucson, Arizona
| | | | - Jessica A Martinez
- Department of Nutritional Sciences, University of Arizona, Tucson, Arizona
- University of Arizona Cancer Center, Tucson, Arizona
| | - Kirsten H Limesand
- Department of Nutritional Sciences, University of Arizona, Tucson, Arizona
- University of Arizona Cancer Center, Tucson, Arizona
| | - Megha Padi
- Bioinformatics Shared Resource, Arizona Cancer Center, University of Arizona, Tucson, Arizona
- University of Arizona Cancer Center, Tucson, Arizona
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona
| |
Collapse
|
14
|
Radiation-Induced Salivary Gland Dysfunction: Mechanisms, Therapeutics and Future Directions. J Clin Med 2020; 9:jcm9124095. [PMID: 33353023 PMCID: PMC7767137 DOI: 10.3390/jcm9124095] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022] Open
Abstract
Salivary glands sustain collateral damage following radiotherapy (RT) to treat cancers of the head and neck, leading to complications, including mucositis, xerostomia and hyposalivation. Despite salivary gland-sparing techniques and modified dosing strategies, long-term hypofunction remains a significant problem. Current therapeutic interventions provide temporary symptom relief, but do not address irreversible glandular damage. In this review, we summarize the current understanding of mechanisms involved in RT-induced hyposalivation and provide a framework for future mechanistic studies. One glaring gap in published studies investigating RT-induced mechanisms of salivary gland dysfunction concerns the effect of irradiation on adjacent non-irradiated tissue via paracrine, autocrine and direct cell-cell interactions, coined the bystander effect in other models of RT-induced damage. We hypothesize that purinergic receptor signaling involving P2 nucleotide receptors may play a key role in mediating the bystander effect. We also discuss promising new therapeutic approaches to prevent salivary gland damage due to RT.
Collapse
|
15
|
Jensen SB, Vissink A, Limesand KH, Reyland ME. Salivary Gland Hypofunction and Xerostomia in Head and Neck Radiation Patients. J Natl Cancer Inst Monogr 2020; 2019:5551361. [PMID: 31425600 DOI: 10.1093/jncimonographs/lgz016] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The most manifest long-term consequences of radiation therapy in the head and neck cancer patient are salivary gland hypofunction and a sensation of oral dryness (xerostomia). METHODS This critical review addresses the consequences of radiation injury to salivary gland tissue, the clinical management of salivary gland hypofunction and xerostomia, and current and potential strategies to prevent or reduce radiation injury to salivary gland tissue or restore the function of radiation-injured salivary gland tissue. RESULTS Salivary gland hypofunction and xerostomia have severe implications for oral functioning, maintenance of oral and general health, and quality of life. Significant progress has been made to spare salivary gland function chiefly due to advances in radiation techniques. Other strategies have also been developed, e.g., radioprotectors, identification and preservation/expansion of salivary stem cells by stimulation with cholinergic muscarinic agonists, and application of new lubricating or stimulatory agents, surgical transfer of submandibular glands, and acupuncture. CONCLUSION Many advances to manage salivary gland hypofunction and xerostomia induced by radiation therapy still only offer partial protection since they are often of short duration, lack the protective effects of saliva, or potentially have significant adverse effects. Intensity-modulated radiation therapy (IMRT), and its next step, proton therapy, have the greatest potential as a management strategy for permanently preserving salivary gland function in head and neck cancer patients.Presently, gene transfer to supplement fluid formation and stem cell transfer to increase the regenerative potential in radiation-damaged salivary glands are promising approaches for regaining function and/or regeneration of radiation-damaged salivary gland tissue.
Collapse
Affiliation(s)
- Siri Beier Jensen
- Department of Dentistry and Oral Health, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Arjan Vissink
- Department of Oral and Maxillofacial Surgery, University of Groningen, University Medical Center, Groningen, The Netherlands
| | | | - Mary E Reyland
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
16
|
A hepatocyte growth factor/MET-induced antiapoptotic pathway protects against radiation-induced salivary gland dysfunction. Radiother Oncol 2019; 138:9-16. [DOI: 10.1016/j.radonc.2019.05.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/07/2019] [Accepted: 05/10/2019] [Indexed: 01/05/2023]
|
17
|
Gilman KE, Camden JM, Klein RR, Zhang Q, Weisman GA, Limesand KH. P2X7 receptor deletion suppresses γ-radiation-induced hyposalivation. Am J Physiol Regul Integr Comp Physiol 2019; 316:R687-R696. [PMID: 30892913 DOI: 10.1152/ajpregu.00192.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Head and neck cancer treatments typically involve a combination of surgery and radiotherapy, often leading to collateral damage to nearby tissues causing unwanted side effects. Radiation damage to salivary glands frequently leads to irreversible dysfunction by poorly understood mechanisms. The P2X7 receptor (P2X7R) is a ligand-gated ion channel activated by extracellular ATP released from damaged cells as "danger signals." P2X7R activation initiates apoptosis and is involved in numerous inflammatory disorders. In this study, we utilized P2X7R knockout (P2X7R-/-) mice to determine the role of the receptor in radiation-induced salivary gland damage. Results indicate a dose-dependent increase in γ-radiation-induced ATP release from primary parotid gland cells of wild-type but not P2X7R-/- mice. Despite these differences, apoptosis levels are similar in parotid glands of wild-type and P2X7R-/- mice 24-72 h after radiation. However, γ-radiation caused elevated prostaglandin E2 (PGE2) release from primary parotid cells of wild-type but not P2X7R-/- mice. To attempt to uncover the mechanism underlying differential PGE2 release, we evaluated the expression and activities of cyclooxygenase and PGE synthase isoforms. There were no consistent trends in these mediators following radiation that could explain the reduction in PGE2 release in P2X7R-/- mice. Irradiated P2X7R-/- mice have stimulated salivary flow rates similar to unirradiated controls, whereas irradiated wild-type mice have significantly decreased salivary flow rates compared with unirradiated controls. Notably, treatment with the P2X7R antagonist A438079 preserves stimulated salivary flow rates in wild-type mice following γ-radiation. These data suggest that P2X7R antagonism is a promising approach for preventing γ-radiation-induced hyposalivation.
Collapse
Affiliation(s)
- Kristy E Gilman
- Department of Nutritional Sciences, The University of Arizona , Tucson, Arizona
| | - Jean M Camden
- Christopher S. Bond Life Sciences Center, Department of Biochemistry, The University of Missouri , Columbia, Missouri
| | - Rob R Klein
- Department of Pathology, The University of Arizona , Tucson, Arizona
| | - Qionghui Zhang
- Department of Nutritional Sciences, The University of Arizona , Tucson, Arizona
| | - Gary A Weisman
- Christopher S. Bond Life Sciences Center, Department of Biochemistry, The University of Missouri , Columbia, Missouri
| | - Kirsten H Limesand
- Department of Nutritional Sciences, The University of Arizona , Tucson, Arizona
| |
Collapse
|
18
|
Shubin AD, Felong TJ, Schutrum BE, Joe DSL, Ovitt CE, Benoit DSW. Encapsulation of primary salivary gland cells in enzymatically degradable poly(ethylene glycol) hydrogels promotes acinar cell characteristics. Acta Biomater 2017; 50:437-449. [PMID: 28039063 PMCID: PMC5455143 DOI: 10.1016/j.actbio.2016.12.049] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 12/06/2016] [Accepted: 12/23/2016] [Indexed: 01/08/2023]
Abstract
Radiation therapy for head and neck cancers leads to permanent xerostomia due to the loss of secretory acinar cells in the salivary glands. Regenerative treatments utilizing primary submandibular gland (SMG) cells show modest improvements in salivary secretory function, but there is limited evidence of salivary gland regeneration. We have recently shown that poly(ethylene glycol) (PEG) hydrogels can support the survival and proliferation of SMG cells as multicellular spheres in vitro. To further develop this approach for cell-based salivary gland regeneration, we have investigated how different modes of PEG hydrogel degradation affect the proliferation, cell-specific gene expression, and epithelial morphology within encapsulated salivary gland spheres. Comparison of non-degradable, hydrolytically-degradable, matrix metalloproteinase (MMP)-degradable, and mixed mode-degradable hydrogels showed that hydrogel degradation by any mechanism is required for significant proliferation of encapsulated cells. The expression of acinar phenotypic markers Aqp5 and Nkcc1 was increased in hydrogels that are MMP-degradable compared with other hydrogel compositions. However, expression of secretory acinar proteins Mist1 and Pip was not maintained to the same extent as phenotypic markers, suggesting changes in cell function upon encapsulation. Nevertheless, MMP- and mixed mode-degradability promoted organization of polarized cell types forming tight junctions and expression of the basement membrane proteins laminin and collagen IV within encapsulated SMG spheres. This work demonstrates that cellularly remodeled hydrogels can promote proliferation and gland-like organization by encapsulated salivary gland cells as well as maintenance of acinar cell characteristics required for regenerative approaches. Investigation is required to identify approaches to further enhance acinar secretory properties. STATEMENT OF SIGNIFICANCE Regenerative strategies to replace damaged salivary glands require the function and organization of acinar cells. Hydrogel-based approaches have shown promise to control cell function and phenotype. However, little is known about how specific parameters, such as the mechanism of hydrogel degradation (e.g., hydrolytic or enzymatic), influence the viability, proliferation, organization, and phenotype of salivary gland cells. In this work, it is shown that hydrogel-encapsulated primary salivary gland cell proliferation is dependent upon hydrogel degradation. Hydrogels crosslinked with enzymatically degradable peptides promoted the expression of critical acinar cell markers, which are typically downregulated in primary cultures. Furthermore, salivary gland cells encapsulated in enzymatically- but not hydrolytically-degradable hydrogels displayed highly organized and polarized salivary gland cell markers, which mimics characteristics found in native gland tissue. In sum, results indicate that salivary gland cells respond to cellularly remodeled hydrogels, resulting in self-assembly and organization akin to acini substructures of the salivary gland.
Collapse
Affiliation(s)
- Andrew D Shubin
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Timothy J Felong
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Brittany E Schutrum
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Debria S L Joe
- Department of Biology, Xavier University of Louisiana, New Orleans, LA, United States
| | - Catherine E Ovitt
- Center for Oral Biology, University of Rochester, Rochester, NY, United States; Department of Biomedical Genetics, University of Rochester, Rochester, NY, United States.
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States; Department of Biomedical Genetics, University of Rochester, Rochester, NY, United States; Department of Chemical Engineering, University of Rochester, Rochester, NY, United States; Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States.
| |
Collapse
|
19
|
Meyer S, Chibly AM, Burd R, Limesand KH. Insulin-Like Growth Factor-1-Mediated DNA Repair in Irradiated Salivary Glands Is Sirtuin-1 Dependent. J Dent Res 2016; 96:225-232. [PMID: 28106504 DOI: 10.1177/0022034516677529] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Ionizing radiation is one of the most common cancer treatments; however, the treatment leads to a wide range of debilitating side effects. In patients with head and neck cancer (HNC), the surrounding normal salivary gland is extremely sensitive to therapeutic radiation, and damage to this tissue results in various oral complications and decreased quality of life (QOL). In the current study, mice treated with targeted head and neck radiation showed a significant increase in double-stranded breaks (DSB) in the DNA of parotid salivary gland cells immediately after treatment, and this remained elevated 3 h posttreatment. In contrast, mice pretreated with insulin-like growth factor-1 (IGF-1) showed resolution of the same amount of initial DNA damage by 3 h posttreatment. At acute time points (30 min to 2 h), irradiated parotid glands had significantly decreased levels of the histone deactylase Sirtuin-1 (SirT-1) which has been previously shown to function in DNA repair. Pretreatment with IGF-1 increased SirT-1 protein levels and increased deacetylation of SirT-1 targets involved in DNA repair. Pharmacological inhibition of SirT-1 activity decreased the IGF-1-mediated resolution of DSB. These data suggest that IGF-1 promotes DNA repair in irradiated parotid glands through the maintenance and activation of SirT-1.
Collapse
Affiliation(s)
- S Meyer
- 1 Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA
| | - A M Chibly
- 1 Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA
| | - R Burd
- 1 Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA
| | - K H Limesand
- 1 Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
20
|
Zeidan YH, Xiao N, Cao H, Kong C, Le QT, Sirjani D. Botulinum Toxin Confers Radioprotection in Murine Salivary Glands. Int J Radiat Oncol Biol Phys 2015; 94:1190-7. [PMID: 26907915 DOI: 10.1016/j.ijrobp.2015.12.371] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 12/19/2015] [Accepted: 12/22/2015] [Indexed: 11/17/2022]
Abstract
PURPOSE Xerostomia is a common radiation sequela, which has a negative impact on the quality of life of patients with head and neck cancer. Current treatment strategies offer only partial relief. Botulinum toxins (BTX) have been successfully used in treating a variety of radiation sequelae such as cystitis, proctitis, fibrosis, and facial pain. The purpose of this study was to evaluate the effect of BTX on radiation-induced salivary gland damage. METHODS AND MATERIALS We used a previously established model for murine salivary gland irradiation (IR). The submandibular glands (SMGs) of C5BL/6 mice (n=6/group) were injected with saline or BTX 72 hours before receiving 15 Gy of focal irradiation. Saliva flow was measured 3, 7, and 28 days after treatment. The SMGs were collected for immunohistochemistry, confocal microscopy, and Western blotting. A cytokine array consisting of 40 different mouse cytokines was used to evaluate cytokine profiles after radiation treatment. RESULTS Irradiated mice showed a 50% reduction in saliva flow after 3 days, whereas mice preinjected with BTX had 25% reduction in saliva flow (P<.05). Cell death detected by TUNEL staining was similar in SMG sections of both groups. However, neutrophil infiltrate, detected by myeloperoxidase staining, was 3-fold lower for the BTX treated mice. A cytokine array showed a 2-fold upregulation of LPS-induced chemokine (LIX/CXCL5) 3 days after IR. BTX pretreatment reduced LIX levels by 40%. At 4 weeks after IR, the saline (control) group showed a 40% reduction in basal SMG weight, compared with 20% in the BTX group. Histologically, BTX-pretreated glands showed relative preservation of acinar structures after radiation. CONCLUSIONS These data suggest that BTX pretreatment ameliorates radiation-induced saliva dysfunction. Moreover, we demonstrate a novel role for CXCL5 in the acute phase of salivary gland damage after radiation. These results carry important clinical implications for the treatment of xerostomia in patients with head and neck cancer.
Collapse
Affiliation(s)
- Youssef H Zeidan
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, Florida.
| | - Nan Xiao
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Hongbin Cao
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Christina Kong
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California; Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Davud Sirjani
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
21
|
Acauan MD, Figueiredo MAZ, Cherubini K, Gomes APN, Salum FG. Radiotherapy-induced salivary dysfunction: Structural changes, pathogenetic mechanisms and therapies. Arch Oral Biol 2015; 60:1802-10. [DOI: 10.1016/j.archoralbio.2015.09.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 09/18/2015] [Accepted: 09/20/2015] [Indexed: 01/01/2023]
|
22
|
Acauan MD, Gomes APN, Braga-Filho A, de Figueiredo MAZ, Cherubini K, Salum FG. Effect of low-level laser therapy on irradiated parotid glands--study in mice. JOURNAL OF BIOMEDICAL OPTICS 2015; 20:108002. [PMID: 26502234 DOI: 10.1117/1.jbo.20.10.108002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 09/23/2015] [Indexed: 06/05/2023]
Abstract
The objective of this study was to evaluate the effect of low-level laser therapy (LLLT) on radiotherapy-induced morphological changes and caspase-3 immunodetection in parotids of mice. Forty-one Swiss mice were divided into control, radiotherapy, 2- and 4-J laser groups. The experimental groups were exposed to ionizing radiation in a single session of 10 Gy. In the laser groups, a GaAlAs laser (830 nm, 100 mW, 0.028 cm2, 3.57 W/cm2) was used on the region corresponding to the parotid glands, with 2-J energy (20 s, 71 J/cm2) or 4 J (40 s, 135 J/cm2) per point. LLLT was performed immediately before and 24 h after radiotherapy. One point was applied in each parotid gland. The animals were euthanized 48 h or 7 days after radiotherapy and parotid glands were dissected for morphological analysis and immunodetection of caspase-3. There was no significant difference between groups in the immunodetection of caspase-3, but the laser groups had a lower percentage compared to the radiotherapy group. LLLT promoted the preservation of acinar structure, reduced the occurrence of vacuolation, and stimulated parotid gland vascularization. Of the two LLLT protocols, the one using 4 J of energy showed better results.
Collapse
Affiliation(s)
- Monique Dossena Acauan
- Pontifical Catholic University of Rio Grande do Sul-PUCRS, Oral Medicine Division, São Lucas Hospital, Avenue Ipiranga, 6690, Room 231, CEP: 90610-000, Porto Alegre, RS, Brazil
| | - Ana Paula Neutziling Gomes
- Federal University of Pelotas-UFPEL, Oral Pathology Division, Rua Gonçalves Chaves 457, CEP: 96015-560, Pelotas, RS, Brazil
| | - Aroldo Braga-Filho
- Pontifical Catholic University of Rio Grande do Sul-PUCRS, Radiotherapy Division, São Lucas Hospital, Brazil
| | - Maria Antonia Zancanaro de Figueiredo
- Pontifical Catholic University of Rio Grande do Sul-PUCRS, Oral Medicine Division, São Lucas Hospital, Avenue Ipiranga, 6690, Room 231, CEP: 90610-000, Porto Alegre, RS, Brazil
| | - Karen Cherubini
- Pontifical Catholic University of Rio Grande do Sul-PUCRS, Oral Medicine Division, São Lucas Hospital, Avenue Ipiranga, 6690, Room 231, CEP: 90610-000, Porto Alegre, RS, Brazil
| | - Fernanda Gonçalves Salum
- Pontifical Catholic University of Rio Grande do Sul-PUCRS, Oral Medicine Division, São Lucas Hospital, Avenue Ipiranga, 6690, Room 231, CEP: 90610-000, Porto Alegre, RS, Brazil
| |
Collapse
|
23
|
Cicenas J, Kalyan K, Sorokinas A, Stankunas E, Levy J, Meskinyte I, Stankevicius V, Kaupinis A, Valius M. Roscovitine in cancer and other diseases. ANNALS OF TRANSLATIONAL MEDICINE 2015. [PMID: 26207228 DOI: 10.3978/j.issn.2305-5839.2015.03.61] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Roscovitine [CY-202, (R)-Roscovitine, Seliciclib] is a small molecule that inhibits cyclin-dependent kinases (CDKs) through direct competition at the ATP-binding site. It is a broad-range purine inhibitor, which inhibits CDK1, CDK2, CDK5 and CDK7, but is a poor inhibitor for CDK4 and CDK6. Roscovitine is widely used as a biological tool in cell cycle, cancer, apoptosis and neurobiology studies. Moreover, it is currently evaluated as a potential drug to treat cancers, neurodegenerative diseases, inflammation, viral infections, polycystic kidney disease and glomerulonephritis. This review focuses on the use of roscovitine in the disease model as well as clinical model research.
Collapse
Affiliation(s)
- Jonas Cicenas
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Karthik Kalyan
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Aleksandras Sorokinas
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Edvinas Stankunas
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Josh Levy
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Ingrida Meskinyte
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Vaidotas Stankevicius
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Algirdas Kaupinis
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| | - Mindaugas Valius
- 1 CALIPHO Group, Swiss Institute of Bioinformatics, Geneva, Switzerland ; 2 MAP Kinase Resource, Bern, Switzerland ; 3 Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius, Lithuania ; 4 Systems Biomedicine Division and Department of Virology and Immunology, Haffkine Institute for Training Research and Testing, Mumbai, India ; 5 Department of Biochemistry, Vilnius University, Vilnius, Lithuania ; 6 RTI International, Research Triangle Park, NC, USA ; 7 Lithuanian Centre of Non-Formal Youth Education Vilnius, Lithuania ; 8 National Cancer Institute, Vilnius, Lithuania ; 9 Vilnius University, Vilnius, Lithuania
| |
Collapse
|
24
|
Pharmacological activation of the EDA/EDAR signaling pathway restores salivary gland function following radiation-induced damage. PLoS One 2014; 9:e112840. [PMID: 25409170 PMCID: PMC4237357 DOI: 10.1371/journal.pone.0112840] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 10/17/2014] [Indexed: 02/06/2023] Open
Abstract
Radiotherapy of head and neck cancers often results in collateral damage to adjacent salivary glands associated with clinically significant hyposalivation and xerostomia. Due to the reduced capacity of salivary glands to regenerate, hyposalivation is treated by substitution with artificial saliva, rather than through functional restoration of the glands. During embryogenesis, the ectodysplasin/ectodysplasin receptor (EDA/EDAR) signaling pathway is a critical element in the development and growth of salivary glands. We have assessed the effects of pharmacological activation of this pathway in a mouse model of radiation-induced salivary gland dysfunction. We report that post-irradiation administration of an EDAR-agonist monoclonal antibody (mAbEDAR1) normalizes function of radiation damaged adult salivary glands as determined by stimulated salivary flow rates. In addition, salivary gland structure and homeostasis is restored to pre-irradiation levels. These results suggest that transient activation of pathways involved in salivary gland development could facilitate regeneration and restoration of function following damage.
Collapse
|
25
|
Cicenas J, Kalyan K, Sorokinas A, Jatulyte A, Valiunas D, Kaupinis A, Valius M. Highlights of the Latest Advances in Research on CDK Inhibitors. Cancers (Basel) 2014; 6:2224-42. [PMID: 25349887 PMCID: PMC4276963 DOI: 10.3390/cancers6042224] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/30/2014] [Accepted: 10/14/2014] [Indexed: 11/16/2022] Open
Abstract
Uncontrolled proliferation is the hallmark of cancer and other proliferative disorders and abnormal cell cycle regulation is, therefore, common in these diseases. Cyclin-dependent kinases (CDKs) play a crucial role in the control of the cell cycle and proliferation. These kinases are frequently deregulated in various cancers, viral infections, neurodegenerative diseases, ischemia and some proliferative disorders. This led to a rigorous pursuit for small-molecule CDK inhibitors for therapeutic uses. Early efforts to block CDKs with nonselective CDK inhibitors led to little specificity and efficacy but apparent toxicity, but the recent advance of selective CDK inhibitors allowed the first successful efforts to target these kinases for the therapies of several diseases. Major ongoing efforts are to develop CDK inhibitors as monotherapies and rational combinations with chemotherapy and other targeted drugs.
Collapse
Affiliation(s)
- Jonas Cicenas
- CALIPHO Group, Swiss Institute of Bioinformatics, CMU-1, rue Michel Servet' Geneva 4 CH-1211, Switzerland.
| | | | | | | | | | - Algirdas Kaupinis
- Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius LT-08662, Lithuania.
| | - Mindaugas Valius
- Proteomics Centre, Vilnius University Institute of Biochemistry, Vilnius LT-08662, Lithuania.
| |
Collapse
|
26
|
Vissink A, Luijk P, Langendijk JA, Coppes RP. Current ideas to reduce or salvage radiation damage to salivary glands. Oral Dis 2014; 21:e1-10. [DOI: 10.1111/odi.12222] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 12/23/2013] [Accepted: 12/23/2013] [Indexed: 12/14/2022]
Affiliation(s)
- A Vissink
- Department of Oral and Maxillofacial Surgery University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - P Luijk
- Department of Oral and Maxillofacial Surgery University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - JA Langendijk
- Department of Radiation Oncology University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - RP Coppes
- Department of Radiation Oncology University of Groningen University Medical Center Groningen Groningen The Netherlands
- Department of Cell Biology Section of Radiation and Stress Biology University of Groningen University Medical Center Groningen Groningen The Netherlands
| |
Collapse
|
27
|
Chibly AM, Nguyen T, Limesand KH. Palliative Care for Salivary Gland Dysfunction Highlights the Need for Regenerative Therapies: A Review on Radiation and Salivary Gland Stem Cells. ACTA ACUST UNITED AC 2014; 4. [PMID: 26693098 DOI: 10.4172/2165-7386.1000180] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Radiotherapy remains the major course of treatment for Head and Neck cancer patients. A common consequence of radiation treatment is dysfunction of the salivary glands, which leads to a number of oral complications including xerostomia and dysphagia, for which there is no existent cure. Here, we briefly describe the current palliative treatments available for patients undergoing these conditions, such as oral lubricants, saliva substitutes, and saliva stimulants. None of these options achieves restoration of normal quality of life due to their limited effectiveness, and in some cases, adverse side effects of their own. Other therapies under development, such as acupuncture and electrostimulation have also yielded mixed results in clinical trials. Due to the ineffectiveness of palliative care to restore quality of life, it is reasonable to aim for the development of regenerative therapies that allow restoration of function of the salivary epithelium following radiation treatment. Adult stem cells are a necessary component of wound healing, and play important roles in preserving normal function of adult tissues. Thus, the present review mainly focuses on the effects of radiation on adult stem cells in a variety of tissues, which may be at play in the response of salivary glands to radiation treatment. This is of clinical importance because progenitor cells of the salivary glands have shown partial regenerative potential in mouse transplantation assays. Therefore, understanding how these progenitor cells are affected by radiation offers potential for development of new therapies for patients with xerostomia.
Collapse
Affiliation(s)
| | - Thao Nguyen
- The University of Arizona Nutritional Sciences Graduate Program, Tucson, AZ 85721, USA
| | - Kirsten H Limesand
- The University of Arizona Nutritional Sciences Graduate Program, Tucson, AZ 85721, USA
| |
Collapse
|
28
|
Morgan-Bathke M, Lin HH, Chibly AM, Zhang W, Sun X, Chen CH, Flodby P, Borok Z, Wu R, Arnett D, Klein RR, Ann DK, Limesand KH. Deletion of ATG5 shows a role of autophagy in salivary homeostatic control. J Dent Res 2013; 92:911-7. [PMID: 23884556 DOI: 10.1177/0022034513499350] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Autophagy is a catabolic pathway utilized to maintain a balance among the synthesis, degradation, and recycling of cellular components, thereby playing a role in cell growth, development, and homeostasis. Previous studies revealed that a conditional knockout of essential member(s) of autophagy in a variety of tissues causes changes in structure and function of these tissues. Acinar cell-specific expression of knocked-in Cre recombinase through control of aquaporin 5 (Aqp5) promoter/enhancer (Aqp5-Cre) allows us to specifically inactivate Atg5, a protein necessary for autophagy, in salivary acinar cells of Atg5(f/f);Aqp5-Cre mice. There was no difference in apoptotic or proliferation levels in salivary glands of Atg5/Cre mice from each genotype. However, H&E staining and electron microscopy studies revealed modestly enlarged acinar cells and accumulated secretory granules in salivary glands of Atg5(f/f);Aqp5-Cre mice. Salivary flow rates and amylase contents of Atg5/Cre mice indicated that acinar-specific inactivation of ATG5 did not alter carbachol-evoked saliva and amylase secretion. Conversely, autophagy intersected with salivary morphological and secretory manifestations induced by isoproterenol administration. These results identified a role for autophagy as a homeostasis control in salivary glands. Collectively, Atg5(f/f);Aqp5-Cre mice would be a useful tool to enhance our understanding of autophagy in adaptive responses following targeted head and neck radiation or Sjögren syndrome.
Collapse
Affiliation(s)
- M Morgan-Bathke
- Department of Nutritional Sciences, The University of Arizona, Tucson, AZ 85721, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Galons H, Oumata N, Gloulou O, Meijer L. Cyclin-dependent kinase inhibitors closer to market launch? Expert Opin Ther Pat 2013; 23:945-63. [PMID: 23600454 DOI: 10.1517/13543776.2013.789861] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Interest in cyclin-dependent kinase (CDK) inhibitors was stimulated by the demonstration that their pharmacological activities could lead to therapies for numerous diseases. Until now, despite the clinical introduction of a dozen compounds belonging to other classes of kinase inhibitors, no CDK inhibitor has reached the marketplace. AREAS COVERED This review covers CDK inhibitor patents published between 2009 and September 2012. It presents compounds currently undergoing clinical development, along with our earlier (2010) review of the same topic, as well as descriptions of recently published compounds not disclosed in the patent literature. It provides the reader with an update of all chemical structures of current interest in the CDK inhibitor field. EXPERT OPINION Though cancer remains the most obvious application for CDK inhibition, other indications, such as HIV infection, could potentially be treated with CDK inhibitors.
Collapse
Affiliation(s)
- Hervé Galons
- Laboratoire de Chimie Organique 2, INSERM U 1022, Université Paris - Descartes, 4 avenue de l'Observatoire, 75270 Paris cedex 06, France.
| | | | | | | |
Collapse
|