1
|
Chen W, Johnston IN. Meta-analyses of executive function deficits in chemotherapy-treated rodent models. Neurosci Biobehav Rev 2025; 173:106131. [PMID: 40194612 DOI: 10.1016/j.neubiorev.2025.106131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/06/2025] [Accepted: 03/28/2025] [Indexed: 04/09/2025]
Abstract
People diagnosed with cancer who undergo chemotherapy commonly encounter cognitive changes, particularly in executive functions (EFs). EFs support goal-directed behaviours, with EF deficits implicated in various neurocognitive impairments. We conducted five meta-analyses of the rodent models to investigate the impact of chemotherapy across five EF domains. A systematic search across PubMed, Web of Science, Scopus, and PsycINFO yielded 56 eligible papers. Our findings supported the clinical literature suggesting the selective impact of chemotherapy on different EF domains. Specifically, chemotherapy-treated animals performed significantly more poorly than controls in tasks assessing working memory, behavioural flexibility, and problem-solving, with no significant group differences in inhibition or attention. Subgroup analyses revealed that alkylating agents, antitumor antibiotics, and combination therapies were strongly associated with working memory deficits, whereas mitotic inhibitors were not. Rodent species, strain, age, sex, number of treatments, and time of behavioural assessment since the end of treatment did not moderate the drug effect on any assessed EF domains. To increase the generalisability and translational validity of the results, the overall reporting quality of animal studies needs to be improved with more details on randomisation, blinding, sample sizes, and criteria for animal exclusions.
Collapse
Affiliation(s)
- Weiye Chen
- School of Psychology, University of Sydney, NSW 2006, Australia
| | - Ian N Johnston
- School of Psychology, University of Sydney, NSW 2006, Australia.
| |
Collapse
|
2
|
Jannapu Reddy S, Mutalik S, Viswanatha GL, Kumar G, John J, Chamallamudi MR, Das A, Das S, Nandakumar K. Nose-to-brain Drug Delivery System: An Emerging Approach to Chemotherapy-induced Cognitive Impairment. Pharm Nanotechnol 2025; 13:212-238. [PMID: 38757164 DOI: 10.2174/0122117385291482240426101519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/26/2024] [Accepted: 03/12/2024] [Indexed: 05/18/2024]
Abstract
The rise in global cancer burden, notably breast cancer, emphasizes the need to address chemotherapy-induced cognitive impairment, also known as chemobrain. Although chemotherapy drugs are effective against cancer, they can trigger cognitive deficits. This has triggered the exploration of preventive strategies and novel therapeutic approaches. Nanomedicine is evolving as a promising tool to be used for the mitigation of chemobrain by overcoming the blood-brain barrier (BBB) with innovative drug delivery systems. Polymer and lipid-based nanoparticles enable targeted drug release, enhancing therapeutic effectiveness. Utilizing the intranasal route of administration may facilitate drug delivery to the central nervous system (CNS) by circumventing first-pass metabolism. Therefore, knowledge of nasal anatomy is critical for optimizing drug delivery via various pathways. Despite challenges, nanoformulations exhibit the potential in enhancing brain drug delivery. Continuous research into formulation techniques and chemobrain mechanisms is vital for developing effective treatments. The intranasal administration of nanoformulations holds promise for improving therapeutic outcomes in chemobrain management. This review offers insights into potential future research directions, such as exploring novel drug combinations, investigating alternative delivery routes, or integrating emerging technologies to enhance the efficacy and safety of nanoformulations for chemobrain management.
Collapse
Affiliation(s)
- Shireesha Jannapu Reddy
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | | | - Gautam Kumar
- Department of Pharmacy, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201310, India
| | - Jeena John
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Mallikarjuna Rao Chamallamudi
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Arpita Das
- Department of Biotechnology, Adamas University, Barasat, Kolkata, 700126, West Bengal, India
| | - Sudip Das
- College of Pharmacy and Health Sciences, Butler University, 4600 Sunset Avenue, Indianapolis, IN 46208, United States
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
3
|
Oppegaard KR, Mayo SJ, Armstrong TS, Dokiparthi V, Melisko M, Levine JD, Olshen AB, Anguera JA, Roy R, Paul S, Cooper B, Conley YP, Hammer MJ, Miaskowski C, Kober KM. Neurodegenerative disease pathways are perturbed in patients with cancer who self-report cognitive changes and anxiety: A pathway impact analysis. Cancer 2024; 130:2834-2847. [PMID: 38676932 DOI: 10.1002/cncr.35336] [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: 08/03/2023] [Revised: 01/31/2024] [Accepted: 03/28/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Cancer-related cognitive impairment (CRCI) and anxiety co-occur in patients with cancer. Little is known about mechanisms for the co-occurrence of these two symptoms. The purposes of this secondary analysis were to evaluate for perturbed pathways associated with the co-occurrence of self-reported CRCI and anxiety in patients with low versus high levels of these two symptoms and to identify potential mechanisms for the co-occurrence of CRCI and anxiety using biological processes common across any perturbed neurodegenerative disease pathways. METHODS Patients completed the Attentional Function Index and the Spielberger State-Trait Anxiety Inventory six times over two cycles of chemotherapy. Based on findings from a previous latent profile analysis, patients were grouped into none versus both high levels of these symptoms. Gene expression was quantified, and pathway impact analyses were performed. Signaling pathways for evaluation were defined with the Kyoto Encyclopedia of Genes and Genomes database. RESULTS A total of 451 patients had data available for analysis. Approximately 85.0% of patients were in the none class and 15.0% were in the both high class. Pathway impact analyses identified five perturbed pathways related to neurodegenerative diseases (i.e., amyotrophic lateral sclerosis, Huntington disease, Parkinson disease, prion disease, and pathways of neurodegeneration-multiple diseases). Apoptosis, mitochondrial dysfunction, oxidative stress, and endoplasmic reticulum stress were common biological processes across these pathways. CONCLUSIONS This study is the first to describe perturbations in neurodegenerative disease pathways associated with CRCI and anxiety in patients receiving chemotherapy. These findings provide new insights into potential targets for the development of mechanistically based interventions.
Collapse
Affiliation(s)
- Kate R Oppegaard
- Department of Physiological Nursing, University of California San Francisco, San Francisco, California, USA
- The Phyllis F. Cantor Center for Research in Nursing and Patient Care Services, Dana-Farber Cancer Institute, University of Massachusetts Boston, Boston, Massachusetts, USA
| | - Samantha J Mayo
- Princess Margaret Cancer Centre, University Health Network, Lawrence S. Bloomberg School of Nursing, University of Toronto, Toronto, Ontario, Canada
| | - Terri S Armstrong
- Neuro-Oncology Branch, Office of Patient-Centered Outcomes Research, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | | | - Michelle Melisko
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jon D Levine
- School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Adam B Olshen
- Department of Epidemiology and Biostatistics, Bakar Computational Health Sciences Institute, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - Joaquin A Anguera
- Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, Sandler Neurosciences Center, University of California San Francisco, San Francisco, California, USA
| | - Ritu Roy
- Helen Diller Family Comprehensive Cancer Center, School of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Steven Paul
- Department of Physiological Nursing, University of California San Francisco, San Francisco, California, USA
| | - Bruce Cooper
- Department of Physiological Nursing, University of California San Francisco, San Francisco, California, USA
| | - Yvette P Conley
- School of Nursing, University of Pittsburg, Pittsburgh, Pennsylvania, USA
| | - Marilyn J Hammer
- The Phyllis F. Cantor Center for Research in Nursing and Patient Care Services, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Christine Miaskowski
- Departments of Physiological Nursing and Anesthesia and Perioperative Care, Pain and Addiction Research Center, University of California San Francisco, San Francisco, California, USA
| | - Kord M Kober
- Department of Physiological Nursing, Bakar Computational Health Sciences Institute, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
4
|
Magdy O, Eshra M, Rashed L, Maher M, Hosny SA, ShamsEldeen AM. Amelioration of cisplatin-induced neurodegenerative changes in rats and restoration of mitochondrial biogenesis by 6-bromoindirubin-3'-oxime: The implication of the GSK-3β/PGC1-α axis. Tissue Cell 2024; 88:102393. [PMID: 38705086 DOI: 10.1016/j.tice.2024.102393] [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: 02/03/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND The cognitive deficits observed after treatment with chemotherapeutic drugs are obvious clinical problems. For treating chemotherapy-induced cognitive deficits (CICD), the treatment modalities must target its underlying mechanisms. Specifically, cisplatin may activate glycogen synthase kinase-3β (GSK-3β), thereby enhancing neuronal apoptosis. 6-bromoindirubin-3'-oxime (6BIO) was not investigated previously in a model of CICD. Therefore, this investigation aimed to address the impacts of GSK3 inhibition on regulating cell signaling, which contributes to neurodegeneration and cognitive impairment. METHODS Thirty adult male Wistar rats were randomly allocated into control groups, while two experimental groups were exposed to repeated cisplatin injections (2 mg/kg intraperitoneally (ip), twice weekly, nine injections), termed chemobrain groups. The rats in the two experimental groups were equally divided into the chemobrain group (untreated) and the chemobrain-6BIO group (treated with 6BIO at a dose of 8.5 μg/kg ip every two days, started after the last dose of cisplatin and continued for two weeks). RESULTS Repeated exposure to cisplatin led to a marked decline in cognitive functions. GSK3 inhibition exerted neuroprotection by decreasing the expression of p-tau and amyloid β, thereby improving cognition. 6BIO, the GSK-3β inhibitor, restored mitochondrial biogenesis by augmenting the protein levels of PGC1-α and increasing the number of mitochondria in the cerebral cortex and hippocampus. CONCLUSION 6BIO provided neuroprotection and exhibited anti-apoptotic and anti-oxidative effects in a rat model of chemobrain.
Collapse
Affiliation(s)
- Ola Magdy
- Department of Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Mohammed Eshra
- Department of Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Laila Rashed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt
| | - Muhammed Maher
- Department of Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Sara Adel Hosny
- Department of Histology, Faculty of Medicine, Cairo University, Egypt
| | | |
Collapse
|
5
|
Lomeli N, Pearre DC, Cruz M, Di K, Ricks-Oddie JL, Bota DA. Cisplatin induces BDNF downregulation in middle-aged female rat model while BDNF enhancement attenuates cisplatin neurotoxicity. Exp Neurol 2024; 375:114717. [PMID: 38336286 PMCID: PMC11087041 DOI: 10.1016/j.expneurol.2024.114717] [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: 07/17/2023] [Revised: 01/04/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
Cancer-related cognitive impairments (CRCI) are neurological complications associated with cancer treatment, and greatly affect cancer survivors' quality of life. Brain-derived neurotrophic factor (BDNF) plays an essential role in neurogenesis, learning and memory. The reduction of BDNF is associated with the decrease in cognitive function in various neurological disorders. Few pre-clinical studies have reported on the effects of chemotherapy and medical stress on BDNF levels and cognition. The present study aimed to compare the effects of medical stress and cisplatin on serum BDNF levels and cognitive function in 9-month-old female Sprague Dawley rats to age-matched controls. Serum BDNF levels were collected longitudinally during cisplatin treatment, and cognitive function was assessed by novel object recognition (NOR) 14 weeks post-cisplatin initiation. Terminal BDNF levels were collected 24 weeks after cisplatin initiation. In cultured hippocampal neurons, we screened three neuroprotective agents, riluzole (an approved treatment for amyotrophic lateral sclerosis), as well as the ampakines CX546 and CX1739. We assessed dendritic arborization by Sholl analysis and dendritic spine density by quantifying postsynaptic density-95 (PSD-95) puncta. Cisplatin and exposure to medical stress reduced serum BDNF levels and impaired object discrimination in NOR compared to age-matched controls. Pharmacological BDNF augmentation protected neurons against cisplatin-induced reductions in dendritic branching and PSD-95. Ampakines (CX546 and CX1739) and riluzole did not affect the antitumor efficacy of cisplatin in vitro. In conclusion, we established the first middle-aged rat model of cisplatin-induced CRCI, assessing the contribution of medical stress and longitudinal changes in BDNF levels on cognitive function, although future studies are warranted to assess the efficacy of BDNF enhancement in vivo on synaptic plasticity. Collectively, our results indicate that cancer treatment exerts long-lasting changes in BDNF levels, and support BDNF enhancement as a potential preventative approach to target CRCI with therapeutics that are FDA approved and/or in clinical study for other indications.
Collapse
Affiliation(s)
- Naomi Lomeli
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - Diana C Pearre
- Gynecologic Oncology, Providence Specialty Medical Group, Burbank, CA, USA
| | - Maureen Cruz
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - Kaijun Di
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - Joni L Ricks-Oddie
- Center for Statistical Consulting, Department of Statistics, University of California Irvine, Irvine, CA, USA; Biostatistics, Epidemiology and Research Design Unit, Institute for Clinical and Translational Sciences, University of California Irvine, Irvine, CA, USA
| | - Daniela A Bota
- Department of Neurology, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
6
|
Zhao Y, Ou M, Hao X, Zhu T. Metabolic change in monocytes and postoperative morbidity after major abdominal surgery in elderly patients: A prospective cohort study. Heliyon 2024; 10:e28137. [PMID: 38571614 PMCID: PMC10987940 DOI: 10.1016/j.heliyon.2024.e28137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/22/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024] Open
Abstract
Background Postoperative complications in aging patients remain a significant cause of increased costs, hospital length of stay, and patient distress. Although alterations in energy metabolism have been closely linked to aging process and surgery, it is still unclear whether metabolic changes during surgery is associated with postoperative complications in elderly patients. This study was conducted to investigate whether metabolic changes during surgery predicts postoperative complications in elderly patients. Methods We conducted a prospective single-center observational cohort study. 244 adults (aged ≥65 years) who were scheduled for elective major non-cardiac surgery were recruited. Blood samples for each patient were taken before and after surgery. All patients were randomly divided into two groups (122 in each group), then oxygen consumption rate (OCR) or extracellular acidification rate (ECAR) was measured on isolated monocytes in each group. Results 14 of 110 (12.7%) patients went through OCR measurement and 15 of 122 patients (12.3%) went through ECAR measurement experienced moderate to severe complications. Overall, there was an intensification of glycolysis in monocytes after surgery. Among all variables, only the change (preoperative -postoperative) of glycolytic reserve (GR)/glycolysis (G) and GR/non-glycolytic acidification (NG) were predictors of moderate to severe complications (AUC = 0.70; 95% CI, 0.56-0.81; P = 0.019 and AUC = 0.67; 95% CI, 0.55-0.80; P = 0.031). Decreased postoperative GR/G were associated with worse postoperative complications (RR = 9.08; 95% CI, 1.23-66.81; P = 0.024). Conclusions Compared with mitochondria function, the change of glycolytic function in monocyte was more valuable in predicting postoperative complications after major abdominal surgery. Our study gave us a new insight into identifying patients at high risk in aging patients.
Collapse
Affiliation(s)
| | | | - Xuechao Hao
- Department of Anesthesiology, and the Research Units of West China (2018RU012) - Chinese Academy of Medical Sciences, West China Hospital of Sichuan University, Sichuan University, Chengdu, China
| | - Tao Zhu
- Department of Anesthesiology, and the Research Units of West China (2018RU012) - Chinese Academy of Medical Sciences, West China Hospital of Sichuan University, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Kim T, James BT, Kahn MC, Blanco-Duque C, Abdurrob F, Islam MR, Lavoie NS, Kellis M, Tsai LH. Gamma entrainment using audiovisual stimuli alleviates chemobrain pathology and cognitive impairment induced by chemotherapy in mice. Sci Transl Med 2024; 16:eadf4601. [PMID: 38446899 PMCID: PMC11588349 DOI: 10.1126/scitranslmed.adf4601] [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: 10/23/2022] [Accepted: 02/15/2024] [Indexed: 03/08/2024]
Abstract
Patients with cancer undergoing chemotherapy frequently experience a neurological condition known as chemotherapy-related cognitive impairment, or "chemobrain," which can persist for the remainder of their lives. Despite the growing prevalence of chemobrain, both its underlying mechanisms and treatment strategies remain poorly understood. Recent findings suggest that chemobrain shares several characteristics with neurodegenerative diseases, including chronic neuroinflammation, DNA damage, and synaptic loss. We investigated whether a noninvasive sensory stimulation treatment we term gamma entrainment using sensory stimuli (GENUS), which has been shown to alleviate aberrant immune and synaptic pathologies in mouse models of neurodegeneration, could also mitigate chemobrain phenotypes in mice administered a chemotherapeutic drug. When administered concurrently with the chemotherapeutic agent cisplatin, GENUS alleviated cisplatin-induced brain pathology, promoted oligodendrocyte survival, and improved cognitive function in a mouse model of chemobrain. These effects persisted for up to 105 days after GENUS treatment, suggesting the potential for long-lasting benefits. However, when administered to mice 90 days after chemotherapy, GENUS treatment only provided limited benefits, indicating that it was most effective when used to prevent the progression of chemobrain pathology. Furthermore, we demonstrated that the effects of GENUS in mice were not limited to cisplatin-induced chemobrain but also extended to methotrexate-induced chemobrain. Collectively, these findings suggest that GENUS may represent a versatile approach for treating chemobrain induced by different chemotherapy agents.
Collapse
Affiliation(s)
- TaeHyun Kim
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Benjamin T. James
- Computer Science and Artificial intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Martin C. Kahn
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Cristina Blanco-Duque
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Fatema Abdurrob
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Md Rezaul Islam
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nicolas S. Lavoie
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Manolis Kellis
- Computer Science and Artificial intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad institute of MIT and Harvard, Cambridge, MA, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad institute of MIT and Harvard, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
8
|
Mahmoud Janloo Y, Attari FS, Roshan S, Lotfi H, Pezeshki AH, Hosseinzadeh M, Shakiba-Jam B, Kafami M. Effect of hydro-alcoholic extract of Nigella sativa on cisplatin-induced memory impairment and brain oxidative stress status in male rats. AVICENNA JOURNAL OF PHYTOMEDICINE 2024; 14:13-22. [PMID: 38948178 PMCID: PMC11210697 DOI: 10.22038/ajp.2023.22789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 09/11/2022] [Indexed: 07/02/2024]
Abstract
Objective Studies have shown the complications of chemotherapy on learning and memory. Empirical evidence suggests that Nigella sativa (NS) has neuroprotective activities. Therefore, the aim of our study was to investigate the effects of NS on cisplatin-induced memory impairment. Materials and Methods This study was conducted on 40 male rats grouped as: control (saline: 2 ml/kg, intraperitoneally (IP), once weekly/2 weeks), cisplatin (Cis, 2 mg/kg, IP, once weekly/2 weeks), NS (200 mg/kg, IP, once weekly/2 weeks), Cis +NS 200 (2 mg/kg Cis + 200 mg/kg NS, IP, once weekly/2 weeks), and Cis +NS 400 (2 mg/kg Cis + 400 mg/kg NS, IP, once weekly/2 weeks). Morris water maze (MWM) test was used to assess spatial learning and memory. In addition, superoxide dismutase (SOD) activity, and thiol and malondialdehyde (MDA) levels were evaluated in the brain. Results Cis significantly enhanced the traveled distance and time spent in the target quadrant in the MWM test. Additionally, MDA levels increased in the Cis group, while thiol and SOD decreased in this group. As a result of treatment with NS, behavioral results were reversed in the groups receiving NS compared to the Cis group. Also, NS reduced MDA level but improved SOD and thiol levels in brain tissue samples. Conclusion NS could improve memory impairment and oxidative stress in animals receiving Cis. Therefore, NS could be used as a potential food supplement to prevent neurotoxicity in patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Yasin Mahmoud Janloo
- Student Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Fatemeh Sadat Attari
- Student Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Sahar Roshan
- Student Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Hadi Lotfi
- Leishmaniasis Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Amir Hossein Pezeshki
- Department of Pathology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoud Hosseinzadeh
- Student Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Batool Shakiba-Jam
- Student Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Marzieh Kafami
- Non-Communicable Disease Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
- Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| |
Collapse
|
9
|
Khodabakhsh P, Asgari Taei A, Shafaroodi H, Pournajaf S, Dargahi L. Effect of Metformin on Epidermal Neural Crest Stem Cells and Their Potential Application in Ameliorating Paclitaxel-induced Neurotoxicity Phenotype. Stem Cell Rev Rep 2024; 20:394-412. [PMID: 37924435 DOI: 10.1007/s12015-023-10642-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2023] [Indexed: 11/06/2023]
Abstract
AIMS Epidermal Neural Crest Stem Cells (EPI-NCSCs) have emerged as prospective ideal candidates to meet the fundamental requirements of cell-based therapies in neurodegenerative disorders. The present study aimed to identify the potential of metformin in driving EPI-NCSCs to neuronal/glial differentiation and express neurotrophic factors as well as assess their therapeutic potential for mitigating the main behavioral manifestations of chemotherapy-induced neurotoxicity (CIN). MAIN METHODS EPI-NCSCs were extracted from the bulge region of hair follicle. Following expansion, transcript and protein expression profiles of key markers for stemness (Nestin, EGR-1, SOX-2 and 10), neurotrophic activity (BDNF, GDNF, NGF, FGF-2, and IL-6), and neuronal (TUB3, DCX, NRF and NeuN) and glial (PDGFRα, NG2, GFAP, and MBP) differentiation were determined on days 1 and 7 post-treatment with 10 and 100 μM metformin using real time-PCR and immunocytochemistry methods. Then, the in vivo function of metformin-treated stem cells was evaluated in the context of paclitaxel CIN. To do so, thermal hyperalgesia, mechanical allodynia, and spatial learning and memory tests were evaluated by Hotplate, Von Frey, and Morris water maze tests. KEY FINDINGS Our result indicated that exposure of EPI-NCSCs to metformin was associated with progressive decline in stemness markers and enhanced expression levels of several neurotrophic, neuron and oligodendrocyte-specific markers. Further, it was observed that intranasal metformin-treated EPI-NCSCs improved the cognitive impairment, and mechanical and thermal hypersensitivity induced by paclitaxel in rats. SIGNIFICANCE Collectively, we reasoned that metformin pretreatment of EPI-NCSCs might further enhance their therapeutic benefits against CIN.
Collapse
Affiliation(s)
- Pariya Khodabakhsh
- Institute of Physiology, Department Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Afsaneh Asgari Taei
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shafaroodi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Safura Pournajaf
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Liu H, Xu K, He Y, Huang F. Mitochondria in Multi-Directional Differentiation of Dental-Derived Mesenchymal Stem Cells. Biomolecules 2023; 14:12. [PMID: 38275753 PMCID: PMC10813276 DOI: 10.3390/biom14010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/03/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
The pursuit of tissue regeneration has fueled decades of research in regenerative medicine. Among the numerous types of mesenchymal stem cells (MSCs), dental-derived mesenchymal stem cells (DMSCs) have recently emerged as a particularly promising candidate for tissue repair and regeneration. In recent years, evidence has highlighted the pivotal role of mitochondria in directing and orchestrating the differentiation processes of DMSCs. Beyond mitochondrial energy metabolism, the multifaceted functions of mitochondria are governed by the mitochondrial quality control (MQC) system, encompassing biogenesis, autophagy, and dynamics. Notably, mitochondrial energy metabolism not only governs the decision to differentiate but also exerts a substantial influence on the determination of differentiation directions. Furthermore, the MQC system exerts a nuanced impact on the differentiation of DMSCs by finely regulating the quality and mass of mitochondria. The review aims to provide a comprehensive overview of the regulatory mechanisms governing the multi-directional differentiation of DMSCs, mediated by both mitochondrial energy metabolism and the MQC system. We also focus on a new idea based on the analysis of data from many research groups never considered before, namely, DMSC-based regenerative medicine applications.
Collapse
Affiliation(s)
| | | | - Yifan He
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510000, China; (H.L.); (K.X.)
| | - Fang Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510000, China; (H.L.); (K.X.)
| |
Collapse
|
11
|
León-Moreno LC, Reza-Zaldívar EE, Hernández-Sapiéns MA, Villafaña-Estarrón E, García-Martin M, Ojeda-Hernández DD, Matias-Guiu JA, Gomez-Pinedo U, Matias-Guiu J, Canales-Aguirre AA. Mesenchymal Stem Cell-Based Therapies in the Post-Acute Neurological COVID Syndrome: Current Landscape and Opportunities. Biomolecules 2023; 14:8. [PMID: 38275749 PMCID: PMC10813738 DOI: 10.3390/biom14010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
One of the main concerns related to SARS-CoV-2 infection is the symptoms that could be developed by survivors, known as long COVID, a syndrome characterized by persistent symptoms beyond the acute phase of the infection. This syndrome has emerged as a complex and debilitating condition with a diverse range of manifestations affecting multiple organ systems. It is increasingly recognized for affecting the Central Nervous System, in which one of the most prevalent manifestations is cognitive impairment. The search for effective therapeutic interventions has led to growing interest in Mesenchymal Stem Cell (MSC)-based therapies due to their immunomodulatory, anti-inflammatory, and tissue regenerative properties. This review provides a comprehensive analysis of the current understanding and potential applications of MSC-based interventions in the context of post-acute neurological COVID-19 syndrome, exploring the underlying mechanisms by which MSCs exert their effects on neuroinflammation, neuroprotection, and neural tissue repair. Moreover, we discuss the challenges and considerations specific to employing MSC-based therapies, including optimal delivery methods, and functional treatment enhancements.
Collapse
Affiliation(s)
- Lilia Carolina León-Moreno
- Unidad de Evaluación Preclínica, Biotecnología Médica Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara 44270, Mexico; (L.C.L.-M.); (M.A.H.-S.); (E.V.-E.)
| | | | - Mercedes Azucena Hernández-Sapiéns
- Unidad de Evaluación Preclínica, Biotecnología Médica Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara 44270, Mexico; (L.C.L.-M.); (M.A.H.-S.); (E.V.-E.)
| | - Erika Villafaña-Estarrón
- Unidad de Evaluación Preclínica, Biotecnología Médica Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara 44270, Mexico; (L.C.L.-M.); (M.A.H.-S.); (E.V.-E.)
| | - Marina García-Martin
- Laboratorio de Neurobiología, Instituto de Investigación Sanitaria, Hospital Clínico San Carlos, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.G.-M.); (D.D.O.-H.); (J.A.M.-G.); (U.G.-P.)
| | - Doddy Denise Ojeda-Hernández
- Laboratorio de Neurobiología, Instituto de Investigación Sanitaria, Hospital Clínico San Carlos, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.G.-M.); (D.D.O.-H.); (J.A.M.-G.); (U.G.-P.)
| | - Jordi A. Matias-Guiu
- Laboratorio de Neurobiología, Instituto de Investigación Sanitaria, Hospital Clínico San Carlos, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.G.-M.); (D.D.O.-H.); (J.A.M.-G.); (U.G.-P.)
| | - Ulises Gomez-Pinedo
- Laboratorio de Neurobiología, Instituto de Investigación Sanitaria, Hospital Clínico San Carlos, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.G.-M.); (D.D.O.-H.); (J.A.M.-G.); (U.G.-P.)
| | - Jorge Matias-Guiu
- Departamento de Neurología, Instituto de Investigación Sanitaria, Hospital Clínico San Carlos, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Alejandro Arturo Canales-Aguirre
- Unidad de Evaluación Preclínica, Biotecnología Médica Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara 44270, Mexico; (L.C.L.-M.); (M.A.H.-S.); (E.V.-E.)
| |
Collapse
|
12
|
McCully JD, del Nido PJ, Emani SM. Mitochondrial transplantation: the advance to therapeutic application and molecular modulation. Front Cardiovasc Med 2023; 10:1268814. [PMID: 38162128 PMCID: PMC10757322 DOI: 10.3389/fcvm.2023.1268814] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/23/2023] [Indexed: 01/03/2024] Open
Abstract
Mitochondrial transplantation provides a novel methodology for rescue of cell viability and cell function following ischemia-reperfusion injury and applications for other pathologies are expanding. In this review we present our methods and acquired data and evidence accumulated to support the use of mitochondrial transplantation.
Collapse
Affiliation(s)
- James D. McCully
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Pedro J. del Nido
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Sitaram M. Emani
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
13
|
Al-Dhalimy AMB, Salim HM, Shather AH, Naser IH, Hizam MM, Alshujery MK. The pathological and therapeutically role of mesenchymal stem cell (MSC)-derived exosome in degenerative diseases; Particular focus on LncRNA and microRNA. Pathol Res Pract 2023; 250:154778. [PMID: 37683391 DOI: 10.1016/j.prp.2023.154778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/17/2023] [Accepted: 08/20/2023] [Indexed: 09/10/2023]
Abstract
By releasing exosomes, which create the ideal milieu for the resolution of inflammation, mesenchymal stem cells (MSCs) enhance tissue healing and have strong immunomodulatory capabilities. MSCs-derived exosome also can affect tumor progress by a myriad of mechanisms. Exosomes function as a cell-cell communication tool to affect cellular activity in recipient cells and include an array of efficient bioactive chemicals. Understanding the fundamental biology of inflammation ablation, tissue homeostasis, and the creation of therapeutic strategies is particularly interested in the horizontal transfer of exosomal long non-coding RNAs (lncRNA) and microRNAs (miRNAs) to recipient cells, where they affect target gene expression. Herein, we propose an exosomal lncRNA and microRNA profile in neurological, renal, cardiac, lung, and liver diseases as well as skin wounds and arthritis.
Collapse
Affiliation(s)
| | - Haitham Mukhlif Salim
- Ministry of Health, Directorat of the Public Health, Health Promotion Departments, Baghdad, Iraq
| | - A H Shather
- Department of Computer Engineering Technology, Al Kitab University, Altun Kopru, Kirkuk 00964, Iraq
| | - Israa Habeeb Naser
- Medical Laboratories Techniques Department, AL-Mustaqbal University, 51001 Hillah, Babil, Iraq
| | - Manar Mohammed Hizam
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | | |
Collapse
|
14
|
Kesler SR, Henneghan AM, Prinsloo S, Palesh O, Wintermark M. Neuroimaging based biotypes for precision diagnosis and prognosis in cancer-related cognitive impairment. Front Med (Lausanne) 2023; 10:1199605. [PMID: 37720513 PMCID: PMC10499624 DOI: 10.3389/fmed.2023.1199605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023] Open
Abstract
Cancer related cognitive impairment (CRCI) is commonly associated with cancer and its treatments, yet the present binary diagnostic approach fails to capture the full spectrum of this syndrome. Cognitive function is highly complex and exists on a continuum that is poorly characterized by dichotomous categories. Advanced statistical methodologies applied to symptom assessments have demonstrated that there are multiple subclasses of CRCI. However, studies suggest that relying on symptom assessments alone may fail to account for significant differences in the neural mechanisms that underlie a specific cognitive phenotype. Treatment plans that address the specific physiologic mechanisms involved in an individual patient's condition is the heart of precision medicine. In this narrative review, we discuss how biotyping, a precision medicine framework being utilized in other mental disorders, could be applied to CRCI. Specifically, we discuss how neuroimaging can be used to determine biotypes of CRCI, which allow for increased precision in prediction and diagnosis of CRCI via biologic mechanistic data. Biotypes may also provide more precise clinical endpoints for intervention trials. Biotyping could be made more feasible with proxy imaging technologies or liquid biomarkers. Large cross-sectional phenotyping studies are needed in addition to evaluation of longitudinal trajectories, and data sharing/pooling is highly feasible with currently available digital infrastructures.
Collapse
Affiliation(s)
- Shelli R. Kesler
- Division of Adult Health, School of Nursing, The University of Texas at Austin, Austin, TX, United States
- Department of Diagnostic Medicine, Dell School of Medicine, The University of Texas at Austin, Austin, TX, United States
- Department of Oncology, Dell School of Medicine, The University of Texas at Austin, Austin, TX, United States
| | - Ashley M. Henneghan
- Division of Adult Health, School of Nursing, The University of Texas at Austin, Austin, TX, United States
- Department of Oncology, Dell School of Medicine, The University of Texas at Austin, Austin, TX, United States
| | - Sarah Prinsloo
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Oxana Palesh
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, United States
| | - Max Wintermark
- Department of Neuroradiology, The University of Texas MD Anderson Cancer, Houston, TX, United States
| |
Collapse
|
15
|
Oliveros A, Poleschuk M, Cole PD, Boison D, Jang MH. Chemobrain: An accelerated aging process linking adenosine A 2A receptor signaling in cancer survivors. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 170:267-305. [PMID: 37741694 PMCID: PMC10947554 DOI: 10.1016/bs.irn.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2023]
Abstract
Chemotherapy has a significant positive impact in cancer treatment outcomes, reducing recurrence and mortality. However, many cancer surviving children and adults suffer from aberrant chemotherapy neurotoxic effects on learning, memory, attention, executive functioning, and processing speed. This chemotherapy-induced cognitive impairment (CICI) is referred to as "chemobrain" or "chemofog". While the underlying mechanisms mediating CICI are still unclear, there is strong evidence that chemotherapy accelerates the biological aging process, manifesting as effects which include telomere shortening, epigenetic dysregulation, oxidative stress, mitochondrial defects, impaired neurogenesis, and neuroinflammation, all of which are known to contribute to increased anxiety and neurocognitive decline. Despite the increased prevalence of CICI, there exists a lack of mechanistic understanding by which chemotherapy detrimentally affects cognition in cancer survivors. Moreover, there are no approved therapeutic interventions for this condition. To address this gap in knowledge, this review attempts to identify how adenosine signaling, particularly through the adenosine A2A receptor, can be an essential tool to attenuate accelerated aging phenotypes. Importantly, the adenosine A2A receptor uniquely stands at the crossroads of cancer treatment and improved cognition, given that it is widely known to control tumor induced immunosuppression in the tumor microenvironment, while also posited to be an essential regulator of cognition in neurodegenerative disease. Consequently, we propose that the adenosine A2A receptor may provide a multifaceted therapeutic strategy to enhance anticancer activity, while combating chemotherapy induced cognitive deficits, both which are essential to provide novel therapeutic interventions against accelerated aging in cancer survivors.
Collapse
Affiliation(s)
- Alfredo Oliveros
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Michael Poleschuk
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Peter D Cole
- Division of Pediatric Hematology/Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, United States.
| | - Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, United States.
| |
Collapse
|
16
|
Jaiswara PK, Shukla SK. Chemotherapy-Mediated Neuronal Aberration. Pharmaceuticals (Basel) 2023; 16:1165. [PMID: 37631080 PMCID: PMC10459787 DOI: 10.3390/ph16081165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/10/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Chemotherapy is a life-sustaining therapeutic option for cancer patients. Despite the advancement of several modern therapies, such as immunotherapy, gene therapy, etc., chemotherapy remains the first-line therapy for most cancer patients. Along with its anti-cancerous effect, chemotherapy exhibits several detrimental consequences that restrict its efficacy and long-term utilization. Moreover, it effectively hampers the quality of life of cancer patients. Cancer patients receiving chemotherapeutic drugs suffer from neurological dysfunction, referred to as chemobrain, that includes cognitive and memory dysfunction and deficits in learning, reasoning, and concentration ability. Chemotherapy exhibits neurotoxicity by damaging the DNA in neurons by interfering with the DNA repair system and antioxidant machinery. In addition, chemotherapy also provokes inflammation by inducing the release of various pro-inflammatory cytokines, including NF-kB, IL-1β, IL-6, and TNF-α. The chemotherapy-mediated inflammation contributes to chemobrain in cancer patients. These inflammatory cytokines modulate several growth signaling pathways and reactive oxygen species homeostasis leading to systemic inflammation in the body. This review is an effort to summarize the available information which discusses the role of chemotherapy-induced inflammation in chemobrain and how it impacts different aspects of therapeutic outcome and the overall quality of life of the patient. Further, this article also discusses the potential of herbal-based remedies to overcome chemotherapy-mediated neuronal toxicity as well as to improve the quality of life of cancer patients.
Collapse
Affiliation(s)
| | - Surendra Kumar Shukla
- Department of Oncology Science, University of Oklahoma Health Science Centre, Oklahoma City, OK 73104, USA;
| |
Collapse
|
17
|
Milutinovic B, Mahalingam R, Mendt M, Arroyo L, Seua A, Dharmaraj S, Shpall E, Heijnen CJ. Intranasally Administered MSC-Derived Extracellular Vesicles Reverse Cisplatin-Induced Cognitive Impairment. Int J Mol Sci 2023; 24:11862. [PMID: 37511623 PMCID: PMC10380450 DOI: 10.3390/ijms241411862] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Neurotoxic side effects of chemotherapy include deficits in attention, memory, and executive functioning. Currently, there are no FDA-approved therapies. In mice, cisplatin causes long-term cognitive deficits, white matter damage, mitochondrial dysfunction, and loss of synaptic integrity. We hypothesized that MSC-derived small extracellular vesicles (sEVs) could restore cisplatin-induced cognitive impairments and brain damage. Animals were injected with cisplatin intraperitoneally and treated with MSC-derived sEVs intranasally 48 and 96 h after the last cisplatin injection. The puzzle box test (PBT) and the novel object place recognition test (NOPRT) were used to determine cognitive deficits. Synaptosomal mitochondrial morphology was analyzed by transmission electron microscopy. Immunohistochemistry using antibodies against synaptophysin and PSD95 was applied to assess synaptic loss. Black-Gold II staining was used to quantify white matter integrity. Our data show that sEVs enter the brain in 30 min and reverse the cisplatin-induced deficits in executive functioning and working and spatial memory. Abnormalities in mitochondrial morphology, loss of white matter, and synaptic integrity in the hippocampus were restored as well. Transcriptomic analysis revealed upregulation of regenerative functions after treatment with sEVs, pointing to a possible role of axonal guidance signaling, netrin signaling, and Wnt/Ca2+ signaling in recovery. Our data suggest that intranasal sEV treatment could become a novel therapeutic approach for the treatment of chemobrain.
Collapse
Affiliation(s)
- Bojana Milutinovic
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rajasekaran Mahalingam
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mayela Mendt
- Department of Stem Cell Transplantation and Cellular Therapy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Luis Arroyo
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alexandre Seua
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shruti Dharmaraj
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cobi J Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
18
|
Scott KS, Chelette B, Chidomere C, Phillip West A, Dantzer R. Cisplatin decreases voluntary wheel-running activity but does not impair food-motivated behavior in mice. Brain Behav Immun 2023; 111:169-176. [PMID: 37076053 PMCID: PMC10330347 DOI: 10.1016/j.bbi.2023.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023] Open
Abstract
Cisplatin is a chemotherapeutic agent that is still commonly used to treat solid tumors. However, it has several toxic side effects due in large part to the mitochondrial damage that it induces. As this mitochondrial damage is likely to result in a decrease in the amount of metabolic energy that is available for behavioral activities, it is not surprising that fatigue develops in cancer patients treated with cisplatin. The present preclinical study was initiated to determine whether the detrimental effects of cisplatin were more pronounced on physical effort requiring a lot of energy versus effort that not only requires less energy but also procures energy in the form of food. For this purpose, mice were either trained to run in a wheel or to work for food in various schedules of food reinforcement before being treated with cisplatin. The experiments were carried out only in male mice as we had already reported that sex differences in cisplatin-induced neurotoxicities are minimal. Cisplatin was administered daily for one cycle of five days, or two cycles separated by a five-day rest. As observed in previous experiments, cisplatin drastically reduced voluntary wheel running. In contrast, when cisplatin was administered to food-restricted mice trained to work for a food reward in a progressive ratio schedule or in a fixed-interval schedule, it tended to increase the number of responses emitted to obtain the food rewards. This increase was not associated with any change in the temporal distribution of responses during the interval between two reinforcements in mice submitted to the fixed interval schedule of food reinforcement. When cisplatin was administered to food-restricted mice trained in an effort-based decision-making task in which they had to choose between working for a grain pellet with little effort and working for a preferred chocolate pellet with more effort, it decreased the total number of responses emitted to obtain food rewards. However, this effect was much less marked than the decrease in wheel running induced by cisplatin. The decrease in the effort invested in the procurement of food rewards was not associated with any change in the relative distribution of effort between low reward and high reward during the time course of the test session. These findings show that cisplatin decreases energy-consuming activities but not energy-procuring activities unless they require a choice between options differing in their cost-benefit ratio. Furthermore, they indicate that the physical dimension of fatigue is more likely to develop in cisplatin-treated individuals than the motivational dimension of fatigue.
Collapse
Affiliation(s)
- Kiersten S Scott
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA
| | - Brandon Chelette
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA
| | - Chinenye Chidomere
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Robert Dantzer
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA.
| |
Collapse
|
19
|
Scott K, Boukelmoune N, Taniguchi C, West AP, Heijnen CJ, Dantzer R. Resolution of cisplatin-induced fatigue does not require endogenous interleukin-10 in male mice. Behav Brain Res 2023; 444:114381. [PMID: 36870396 PMCID: PMC10029095 DOI: 10.1016/j.bbr.2023.114381] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/06/2023]
Abstract
Based on previous results showing a pivotal role of endogenous interleukin-10 (IL-10) in the recovery from cisplatin-induced peripheral neuropathy, the present experiments were carried out to determine whether this cytokine plays any role in the recovery from cisplatin-induced fatigue in male mice. Fatigue was measured by decreased voluntary wheel running in mice trained to run in a wheel in response to cisplatin. Mice were treated with a monoclonal neutralizing antibody (IL-10na) administered intranasally during the recovery period to neutralize endogenous IL-10. In the first experiment, mice were treated with cisplatin (2.83 mg/kg/day) for five days and IL-10na (12 μg/day for three days) five days later. In the second experiment, they were treated with cisplatin (2.3 mg/kg/day for 5 days twice at a five-day interval) and IL10na (12 μg/day for three days) immediately after the last injection of cisplatin. In both experiments, cisplatin decreased body weight and reduced voluntary wheel running. However, IL-10na did not impair recovery from these effects. These results show that the recovery from the cisplatin-induced decrease in wheel running does not require endogenous IL-10 in contrast to the recovery from cisplatin-induced peripheral neuropathy.
Collapse
Affiliation(s)
- Kiersten Scott
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nabila Boukelmoune
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cullen Taniguchi
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX 77087, United States
| | - Cobi J Heijnen
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
20
|
Kinin B1 and B2 Receptors Contribute to Cisplatin-Induced Painful Peripheral Neuropathy in Male Mice. Pharmaceutics 2023; 15:pharmaceutics15030852. [PMID: 36986713 PMCID: PMC10051506 DOI: 10.3390/pharmaceutics15030852] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Cisplatin is the preferential chemotherapeutic drug for highly prevalent solid tumours. However, its clinical efficacy is frequently limited due to neurotoxic effects such as peripheral neuropathy. Chemotherapy-induced peripheral neuropathy is a dose-dependent adverse condition that negatively impacts quality of life, and it may determine dosage limitations or even cancer treatment cessation. Thus, it is urgently necessary to identify pathophysiological mechanisms underlying these painful symptoms. As kinins and their B1 and B2 receptors contribute to the development of chronic painful conditions, including those induced by chemotherapy, the contribution of these receptors to cisplatin-induced peripheral neuropathy was evaluated via pharmacological antagonism and genetic manipulation in male Swiss mice. Cisplatin causes painful symptoms and impaired working and spatial memory. Kinin B1 (DALBK) and B2 (Icatibant) receptor antagonists attenuated some painful parameters. Local administration of kinin B1 and B2 receptor agonists (in sub-nociceptive doses) intensified the cisplatin-induced mechanical nociception attenuated by DALBK and Icatibant, respectively. In addition, antisense oligonucleotides to kinin B1 and B2 receptors reduced cisplatin-induced mechanical allodynia. Thus, kinin B1 and B2 receptors appear to be potential targets for the treatment of cisplatin-induced painful symptoms and may improve patients’ adherence to treatment and their quality of life.
Collapse
|
21
|
Zamorano M, Alexander JF, Catania D, Dharmaraj S, Kavelaars A, Heijnen CJ. Nasal administration of mesenchymal stem cells prevents accelerated age-related tauopathy after chemotherapy in mice. Immun Ageing 2023; 20:5. [PMID: 36698170 PMCID: PMC9874182 DOI: 10.1186/s12979-023-00328-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 01/12/2023] [Indexed: 01/26/2023]
Abstract
BACKGROUND There is increasing concern that cancer and cancer treatment accelerate aging and the associated cognitive decline. We showed recently that treatment of 9-month-old male mice with cisplatin causes cognitive deficits that are associated with formation of tau deposits in the hippocampus. Here we explored the capacity of mesenchymal stem cells (MSC) given via the nose to prevent age-related brain tau deposits. Moreover, we more closely examined the cellular distribution of this hallmark of accelerated brain aging in response to treatment of 9-month-old female and male mice with cisplatin. RESULTS We show that cisplatin induces tau deposits in the entorhinal cortex and hippocampus in both sexes. The tau deposits colocalize with syndecan-2. Astrocytes surrounding tau deposits have increased glial fibrillary acidic protein glial fibrillary acidic protein (GFAP) expression. Most of the cisplatin-induced tau deposits were located in microtubule associated protein-2 (MAP-2)+ neurons that were surrounded by aquaporin 4+ (AQP4)+ neuron-facing membrane domains of astrocytes. In addition, some tau deposits were detected in the perinuclear region of GFAP+ astrocytes and in CD31+ endothelial cells. There were no morphological signs of activation of ionized calcium binding adaptor molecule-1+ (Iba-1)+ microglia and no increases in brain cytokine production. Nasal administration of MSC at 48 and 96 hours after cisplatin prevented formation of tau deposits and normalized syndecan-2 and GFAP expression. Behaviorally, cisplatin-induced tau cluster formation was associated with reduced executive functioning and working/spatial memory and nasal administration of MSC at 48 and 96 hours after cisplatin prevented these cognitive deficits. Notably, delayed MSC administration (1 month after cisplatin) also prevented tau cluster formation and cognitive deficits, in both sexes. CONCLUSION In summary, nasal administration of MSC to older mice at 2 days or 1 month after completion of cisplatin treatment prevents the accelerated development of tau deposits in entorhinal cortex and hippocampus and the associated cognitive deficits. Since MSC are already in clinical use for many other clinical indications, developing nasal MSC administration for treatment of accelerated brain aging and cognitive deficits in cancer survivors should be feasible and would greatly improve their quality of life.
Collapse
Affiliation(s)
- Miriam Zamorano
- grid.240145.60000 0001 2291 4776Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, TX USA ,grid.267308.80000 0000 9206 2401Department of Pediatric Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX USA
| | - Jenolyn F. Alexander
- grid.240145.60000 0001 2291 4776Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, TX USA ,grid.410718.b0000 0001 0262 7331Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr, 55 Essen, Germany
| | - Desiree Catania
- grid.240145.60000 0001 2291 4776Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Shruti Dharmaraj
- grid.240145.60000 0001 2291 4776Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Annemieke Kavelaars
- grid.240145.60000 0001 2291 4776Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Cobi J. Heijnen
- grid.240145.60000 0001 2291 4776Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
22
|
Scott K, Phan TT, Boukelmoune N, Heijnen CJ, Dantzer R. Chronic restraint stress impairs voluntary wheel running but has no effect on food-motivated behavior in mice. Brain Behav Immun 2023; 107:319-329. [PMID: 36349643 PMCID: PMC9729455 DOI: 10.1016/j.bbi.2022.10.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 10/18/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
Abstract
Chronic restraint stress is known to cause significant alterations of mitochondrial biology. However, its effects on effort-based behavior and the sensitivity of these effects to treatments that restore mitochondrial function have not been assessed. Based on the hypothesis that the behavioral consequences of this stressor should be more severe for an energy demanding activity than for an energy procuring activity, we compared the effects of chronic restraint stress on the performance of male mice trained to use a running wheel or to nose poke for a food reward in an operant conditioning cage. In accordance with our hypothesis, we observed that exposure of mice to 2-hour daily restraint sessions for 14 to 16 days during the light phase of the cycle reliably decreased voluntary wheel running but had no effect on working for food in a fixed ratio 10 schedule of food reinforcement or in a progressive ratio schedule of food reinforcement. This dissociation between the two types of behavioral activities could reflect an adaptive response to the constraint imposed by chronic restraint stress on mitochondria function and its negative consequences on energy metabolism. To determine whether it is the case, we administered mesenchymal stem cells intranasally to chronically restrained mice to repair the putative mitochondrial dysfunction induced by chronic restraint stress. This intervention had no effect on wheel running deficits. Assessment of mitochondrial gene expression in the brain of mice submitted to chronic restraint stress revealed an increase in the expression of genes involved in mitochondrial biology that showed habituation with repetition of daily sessions of restraint stress. These original findings can be interpreted to indicate that chronic restraint stress induces behavioral and mitochondrial adjustments that contribute to metabolic adaptation to this stressor and maintain metabolic flexibility.
Collapse
Affiliation(s)
- Kiersten Scott
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Thien Trong Phan
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nabila Boukelmoune
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cobi J Heijnen
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert Dantzer
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
23
|
Savchuk S, Monje M. Mini-Review: Aplastic Myelin Following Chemotherapy. Neurosci Lett 2022; 790:136861. [PMID: 36055447 DOI: 10.1016/j.neulet.2022.136861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/12/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022]
Abstract
The contribution of chemotherapy to improved outcomes for cancer patients is unquestionable. Yet as its applications broaden, so do the concerns for the long-term implications of chemotherapy on the health of cancer survivors, with chemotherapy-related cognitive impairment as a cause for particular urgency. In this mini review, we explore myelin aplasticity following chemotherapy, discussing the role of myelin plasticity in healthy cognition and failure of myelin plasticity chiefly due microenvironmental aberrations in chemotherapy-related cognitive impairment. Possible therapeutic strategies to mitigate chemotherapy-induced myelin dysfunction are also discussed.
Collapse
Affiliation(s)
- Solomiia Savchuk
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA; Department of Neurosurgery, Stanford University, Stanford, CA, 94305, USA; Department of Pathology, Stanford University, Stanford, CA, 94305, USA; Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
24
|
Brain Protection by Methylene Blue and Its Derivative, Azur B, via Activation of the Nrf2/ARE Pathway in Cisplatin-Induced Cognitive Impairment. Pharmaceuticals (Basel) 2022; 15:ph15070815. [PMID: 35890114 PMCID: PMC9320109 DOI: 10.3390/ph15070815] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 02/06/2023] Open
Abstract
Cisplatin is a cytotoxic chemotherapeutic drug that leads to DNA damage and is used in the treatment of various types of tumors. However, cisplatin has several serious adverse effects, such as deterioration in cognitive ability. The aim of our work was to study neuroprotectors capable of preventing cisplatin-induced neurotoxicity. Methylene blue (MB) and AzurB (AzB) are able to neutralize the neurotoxicity caused by cisplatin by protecting nerve cells as a result of the activation of the Ntf2 signaling pathway. We have shown that cisplatin impairs learning in the Morris water maze. This is due to an increase in the amount of mtDNA damage, a decrease in the expression of most antioxidant genes, the main determinant of the induction of which is the Nrf2/ARE signaling pathway, and genes involved in mitophagy regulation in the cortex. The expression of genes involved in long-term potentiation was suppressed in the hippocampus of cisplatin-injected mice. MB in most cases prevented cisplatin-induced impairment of learning and decrease of gene expression in the cortex. AzB prevented the cisplatin-induced decrease of genes in the hippocampus. Also, cisplatin induced disbalance in the gut microbiome, decreased levels of Actinotalea and Prevotella, and increased levels of Streptococcus and Veillonella. MB and AzB also prevented cisplatin-induced changes in the bacterial composition of the gut microbiome.
Collapse
|
25
|
Roy ER, Chiu G, Li S, Propson NE, Kanchi R, Wang B, Coarfa C, Zheng H, Cao W. Concerted type I interferon signaling in microglia and neural cells promotes memory impairment associated with amyloid β plaques. Immunity 2022; 55:879-894.e6. [PMID: 35443157 PMCID: PMC9109419 DOI: 10.1016/j.immuni.2022.03.018] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/02/2021] [Accepted: 03/23/2022] [Indexed: 12/14/2022]
Abstract
The principal signals that drive memory and cognitive impairment in Alzheimer's disease (AD) remain elusive. Here, we revealed brain-wide cellular reactions to type I interferon (IFN-I), an innate immune cytokine aberrantly elicited by amyloid β plaques, and examined their role in cognition and neuropathology relevant to AD in a murine amyloidosis model. Using a fate-mapping reporter system to track cellular responses to IFN-I, we detected robust, Aβ-pathology-dependent IFN-I activation in microglia and other cell types. Long-term blockade of IFN-I receptor (IFNAR) rescued both memory and synaptic deficits and resulted in reduced microgliosis, inflammation, and neuritic pathology. Microglia-specific Ifnar1 deletion attenuated the loss of post-synaptic terminals by selective engulfment, whereas neural Ifnar1 deletion restored pre-synaptic terminals and decreased plaque accumulation. Overall, IFN-I signaling represents a critical module within the neuroinflammatory network of AD and prompts concerted cellular states that are detrimental to memory and cognition.
Collapse
Affiliation(s)
- Ethan R Roy
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gabriel Chiu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sanming Li
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nicholas E Propson
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rupa Kanchi
- Department of Molecular and Cellular Biology and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Baiping Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Wei Cao
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
26
|
Al Dahhan NZ, Cox E, Nieman BJ, Mabbott DJ. Cross-translational models of late-onset cognitive sequelae and their treatment in pediatric brain tumor survivors. Neuron 2022; 110:2215-2241. [PMID: 35523175 DOI: 10.1016/j.neuron.2022.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/21/2022] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
Abstract
Pediatric brain tumor treatments have a high success rate, but survivors are at risk of cognitive sequelae that impact long-term quality of life. We summarize recent clinical and animal model research addressing pathogenesis or evaluating candidate interventions for treatment-induced cognitive sequelae. Assayed interventions encompass a broad range of approaches, including modifications to radiotherapy, modulation of immune response, prevention of treatment-induced cell loss or promotion of cell renewal, manipulation of neuronal signaling, and lifestyle/environmental adjustments. We further emphasize the potential of neuroimaging as a key component of cross-translation to contextualize laboratory research within broader clinical findings. This cross-translational approach has the potential to accelerate discovery to improve pediatric cancer survivors' long-term quality of life.
Collapse
Affiliation(s)
- Noor Z Al Dahhan
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Elizabeth Cox
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada; Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Brian J Nieman
- Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada; Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Donald J Mabbott
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada; Department of Psychology, University of Toronto, Toronto, ON, Canada; Department of Psychology, Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
27
|
Phillips NS, Rao V, Kmetz L, Vela R, Medick S, Krull K, Kesler SR. Changes in Brain Functional and Effective Connectivity After Treatment for Breast Cancer and Implications for Intervention Targets. Brain Connect 2022; 12:385-397. [PMID: 34210168 PMCID: PMC9131353 DOI: 10.1089/brain.2021.0049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: Patients with breast cancer frequently report cognitive impairment both during and after completion of therapy. Evidence suggests that cancer-related cognitive impairments are related to widespread neural network dysfunction. The default mode network (DMN) is a large conserved network that plays a critical role in integrating the functions of various neural systems. Disruption of the network may play a key role in the development of cognitive impairment. Methods: We compared neuroimaging and neurocognitive data from 43 newly diagnosed primary breast cancer patients (mean age = 48, standard deviation [SD] = 8.9 years) and 50 frequency-matched healthy female controls (mean age = 50, SD = 10 years) before treatment and 1 year after treatment completion. Functional and effective connectivity measures of the DMN were obtained using graph theory and Bayesian network analysis methods, respectively. Results: Compared with healthy females, the breast cancer group displayed higher global efficiency and path length post-treatment (p < 0.03, corrected). Breast cancer survivors showed significantly lower performance on measures of verbal memory, attention, and verbal fluency (p < 0.05) at both time points. Within the DMN, local brain network organization, as measured by edge-betweenness centralities, was significantly altered in the breast cancer group compared with controls at both time points (p < 0.0001, corrected), with several connections showing a significant group-by-time effect (p < 0.003, corrected). Effective connectivity demonstrated significantly altered patterns of neuronal coupling in patients with breast cancer (p < 0.05). Significant correlations were seen between hormone blockade therapy, radiation therapy, chemotherapy cycles, memory, and verbal fluency test and edge-betweenness centralities. Discussion: This pattern of altered network organization in the default mode is believed to result in reduced network efficiency and disrupted communication. Subregions of the DMN, the orbital prefrontal cortex and posterior memory network, appear to be at the center of this disruption and this could inform future interventions. Impact statement This prospective study is the first to investigate how post-treatment changes in functional and effective connectivity in the regions of default mode network are related to cancer therapy and measures of memory and verbal learning in breast cancer patients. We demonstrate that the interactions between treatment, brain connectivity, and neurocognitive outcomes coalesce around a subgroup of brain structures in the orbital frontal and parietal lobe. This would suggest that interventions that target these regions may improve neurocognitive outcomes in breast cancer survivors.
Collapse
Affiliation(s)
- Nicholas S. Phillips
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Vikram Rao
- School of Nursing, University of Texas at Austin, Austin, Texas, USA
| | - Lorie Kmetz
- School of Nursing, University of Texas at Austin, Austin, Texas, USA
| | - Ruben Vela
- School of Nursing, University of Texas at Austin, Austin, Texas, USA
- Department of Diagnostic Medicine, Dell School of Medicine, University of Texas at Austin, Austin, Texas, USA
| | - Sarah Medick
- School of Nursing, University of Texas at Austin, Austin, Texas, USA
| | - Kevin Krull
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Shelli R. Kesler
- School of Nursing, University of Texas at Austin, Austin, Texas, USA
- Department of Diagnostic Medicine, Dell School of Medicine, University of Texas at Austin, Austin, Texas, USA
- Center for Computational Oncology, Oden Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
28
|
Alexander JF, Mahalingam R, Seua AV, Wu S, Arroyo LD, Hörbelt T, Schedlowski M, Blanco E, Kavelaars A, Heijnen CJ. Targeting the Meningeal Compartment to Resolve Chemobrain and Neuropathy via Nasal Delivery of Functionalized Mitochondria. Adv Healthc Mater 2022; 11:e2102153. [PMID: 35007407 PMCID: PMC9803615 DOI: 10.1002/adhm.202102153] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/03/2022] [Indexed: 01/03/2023]
Abstract
Cognitive deficits (chemobrain) and peripheral neuropathy occur in ∼75% of patients treated for cancer with chemotherapy and persist long-term in >30% of survivors. Without preventive or curative interventions and with increasing survivorship rates, the population debilitated by these neurotoxicities is rising. Platinum-based chemotherapeutics, including cisplatin, induce neuronal mitochondrial defects leading to chemobrain and neuropathic pain. This study investigates the capacity of nasally administered mesenchymal stem cell-derived mitochondria coated with dextran-triphenylphosphonium polymer (coated mitochondria) to reverse these neurotoxicities. Nasally administered coated mitochondria are rapidly detectable in macrophages in the brain meninges but do not reach the brain parenchyma. The coated mitochondria change expression of >2400 genes regulating immune, neuronal, endocrine and vascular pathways in the meninges of mice treated with cisplatin. Nasal administration of coated mitochondria reverses cisplatin-induced cognitive deficits and resolves neuropathic pain at a >55-times lower dose compared to uncoated mitochondria. Reversal of these neuropathologies is associated with resolution of cisplatin-induced deficits in myelination, synaptosomal mitochondrial integrity and neurogenesis. These findings demonstrate that nasally administered coated mitochondria promote resolution of chemobrain and peripheral neuropathy, thereby identifying a novel facile strategy for clinical application of mitochondrial donation and treating central and peripheral nervous system pathologies by targeting the brain meninges.
Collapse
Affiliation(s)
- Jenolyn F. Alexander
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States,Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, D-45147, Essen, Germany
| | - Rajasekaran Mahalingam
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States
| | - Alexandre V. Seua
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States
| | - Suhong Wu
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas, 77030, United States
| | - Luis D. Arroyo
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States
| | - Tina Hörbelt
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, D-45147, Essen, Germany
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, D-45147, Essen, Germany
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas, 77030, United States
| | - Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States
| | - Cobi J. Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, Texas, 77030, United States,Corresponding Author
| |
Collapse
|
29
|
Sahu K, Singh S, Devi B, Singh C, Singh A. A review on the neuroprotective effect of berberine against chemotherapy-induced cognitive impairment. Curr Drug Targets 2022; 23:913-923. [PMID: 35240956 DOI: 10.2174/1389450123666220303094752] [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: 08/21/2021] [Revised: 12/02/2021] [Accepted: 01/06/2022] [Indexed: 11/22/2022]
Abstract
Chemobrain is one of the major side effects of chemotherapy, despite increased research, the mechanisms underlying chemotherapy-induced cognitive changes remain unknown. Though, several possibly important candidate mechanisms have been identified and will be studied further in the future. Chemobrain is characterized by memory loss, cognitive impairment, difficulty in language, concentration, acceleration, and learning. The major characteristic of chemobrain is oxidative stress, mitochondrial dysfunction, immune dysregulation, hormonal alteration, white matter abnormalities, and DNA damage. Berberine (BBR) is an isoquinoline alkaloid extracted from various berberine species. BBR is a small chemical that easily passes the blood-brain barrier (BBB), making it useful for treating neurodegenerative diseases. Many studies on the pharmacology of BBR have been reported in the past. Furthermore, several clinical and experimental research indicates that BBR has a variety of pharmacological effects. So, in this review, we explore the pathogenesis of chemobrain and the neuroprotective potential of BBR against chemobrain. We also introduced the therapeutic role of BBR in various neurodegenerative and neurological diseases such as Alzheimer's, Parkinson's disease, mental depression, schizophrenia, anxiety, and also some stroke.
Collapse
Affiliation(s)
- Kuleshwar Sahu
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab India
| | - Sukhdev Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab India
| | - Bhawna Devi
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab India
| | - Charan Singh
- Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab-144603, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab India
| |
Collapse
|
30
|
Targeting the A 3 adenosine receptor to prevent and reverse chemotherapy-induced neurotoxicities in mice. Acta Neuropathol Commun 2022; 10:11. [PMID: 35093182 PMCID: PMC8800287 DOI: 10.1186/s40478-022-01315-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
Cisplatin is used to combat solid tumors. However, patients treated with cisplatin often develop cognitive impairments, sensorimotor deficits, and peripheral neuropathy. There is no FDA-approved treatment for these neurotoxicities. We investigated the capacity of a highly selective A3 adenosine receptor (AR) subtype (A3AR) agonist, MRS5980, to prevent and reverse cisplatin-induced neurotoxicities. MRS5980 prevented cisplatin-induced cognitive impairment (decreased executive function and impaired spatial and working memory), sensorimotor deficits, and neuropathic pain (mechanical allodynia and spontaneous pain) in both sexes. At the structural level, MRS5980 prevented the cisplatin-induced reduction in markers of synaptic integrity. In-situ hybridization detected Adora3 mRNA in neurons, microglia, astrocytes and oligodendrocytes. RNAseq analysis identified 164 genes, including genes related to mitochondrial function, of which expression was changed by cisplatin and normalized by MRS5980. Consistently, MRS5980 prevented cisplatin-induced mitochondrial dysfunction and decreased signs of oxidative stress. Transcriptomic analysis showed that the A3AR agonist upregulates genes related to repair pathways including NOTCH1 signaling and chromatin modification in the cortex of cisplatin-treated mice. Importantly, A3AR agonist administration after completion of cisplatin treatment resolved cognitive impairment, neuropathy and sensorimotor deficits. Our results highlight the efficacy of a selective A3AR agonist to prevent and reverse cisplatin-induced neurotoxicities via preventing brain mitochondrial damage and activating repair pathways. An A3AR agonist is already in cancer, clinical trials and our results demonstrate management of neurotoxic side effects of chemotherapy as an additional therapeutic benefit.
Collapse
|
31
|
McAlpin BR, Mahalingam R, Singh AK, Dharmaraj S, Chrisikos TT, Boukelmoune N, Kavelaars A, Heijnen CJ. HDAC6 inhibition reverses long-term doxorubicin-induced cognitive dysfunction by restoring microglia homeostasis and synaptic integrity. Theranostics 2022; 12:603-619. [PMID: 34976203 PMCID: PMC8692908 DOI: 10.7150/thno.67410] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/09/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most common female malignancy in both the developed and developing world. Doxorubicin is one of the most commonly used chemotherapies for breast cancer. Unfortunately, up to 60% of survivors report long-term chemotherapy-induced cognitive dysfunction (CICD) characterized by deficits in working memory, processing speed and executive function. Currently, no therapeutic standard for treating CICD exists. Here, we hypothesized that treatment with a blood-brain barrier permeable histone deacetylase 6 (HDAC6) inhibitor can successfully reverse long-term doxorubicin-induced cognitive dysfunction. Methods: The puzzle box test and novel object/place recognition test were used to assess cognitive function following a therapeutic doxorubicin dosing schedule in female mice. Mitochondrial function and morphology in neuronal synaptosomes were evaluated using the Seahorse XF24 extracellular flux analyzer and transmission electron microscopy, respectively. Hippocampal postsynaptic integrity was evaluated using immunofluorescence. Hippocampal microglia phenotype was determined using advanced imaging techniques and single-nucleus RNA sequencing. Results: A 14-day treatment with a blood-brain barrier permeable HDAC6 inhibitor successfully reversed long-term CICD in the domains of executive function, working and spatial memory. No significant changes in mitochondrial function or morphology in neuronal synaptosomes were detected. Long-term CICD was associated with a decreased expression of postsynaptic PSD95 in the hippocampus. These changes were associated with decreased microglial ramification and alterations in the microglia transcriptome that suggest a stage 1 disease-associated microglia (DAM) phenotype. HDAC6 inhibition completely reversed these doxorubicin-induced alterations, indicating a restoration of microglial homeostasis. Conclusion: Our results show that decreased postsynaptic integrity and a neurodegenerative microglia phenotype closely resembling stage 1 DAM microglia contribute to long-term CICD. Moreover, HDAC6 inhibition shows promise as an efficacious pharmaceutical intervention to alleviate CICD and improve quality of life of breast cancer survivors.
Collapse
Affiliation(s)
- Blake R McAlpin
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rajasekaran Mahalingam
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anand K Singh
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shruti Dharmaraj
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Taylor T Chrisikos
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nabila Boukelmoune
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Cobi J Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- ✉ Corresponding author: Cobi J. Heijnen, Ph.D., 6565 MD Anderson Blvd., Zayed Building Z8.5034, Houston, Texas 77030, Phone 713-563-0162,
| |
Collapse
|
32
|
Perše M. Cisplatin Mouse Models: Treatment, Toxicity and Translatability. Biomedicines 2021; 9:1406. [PMID: 34680523 PMCID: PMC8533586 DOI: 10.3390/biomedicines9101406] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/26/2021] [Accepted: 10/05/2021] [Indexed: 02/06/2023] Open
Abstract
Cisplatin is one of the most widely used chemotherapeutic drugs in the treatment of a wide range of pediatric and adult malignances. However, it has various side effects which limit its use. Cisplatin mouse models are widely used in studies investigating cisplatin therapeutic and toxic effects. However, despite numerous promising results, no significant improvement in treatment outcome has been achieved in humans. There are many drawbacks in the currently used cisplatin protocols in mice. In the paper, the most characterized cisplatin protocols are summarized together with weaknesses that need to be improved in future studies, including hydration and supportive care. As demonstrated, mice respond to cisplatin treatment in similar ways to humans. The paper thus aims to illustrate the complexity of cisplatin side effects (nephrotoxicity, gastrointestinal toxicity, neurotoxicity, ototoxicity and myelotoxicity) and the interconnectedness and interdependence of pathomechanisms among tissues and organs in a dose- and time-dependent manner. The paper offers knowledge that can help design future studies more efficiently and interpret study outcomes more critically. If we want to understand molecular mechanisms and find therapeutic agents that would have a potential benefit in clinics, we need to change our approach and start to treat animals as patients and not as tools.
Collapse
Affiliation(s)
- Martina Perše
- Medical Experimental Centre, Institute of Pathology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
33
|
Ma J, Goodwani S, Acton PJ, Buggia-Prevot V, Kesler SR, Jamal I, Mahant ID, Liu Z, Mseeh F, Roth BL, Chakraborty C, Peng B, Wu Q, Jiang Y, Le K, Soth MJ, Jones P, Kavelaars A, Ray WJ, Heijnen CJ. Inhibition of dual leucine zipper kinase prevents chemotherapy-induced peripheral neuropathy and cognitive impairments. Pain 2021; 162:2599-2612. [PMID: 33872235 PMCID: PMC8442742 DOI: 10.1097/j.pain.0000000000002256] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 01/15/2021] [Accepted: 01/26/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Chemotherapy-induced peripheral neuropathy (CIPN) and chemotherapy-induced cognitive impairments (CICI) are common, often severe neurotoxic side effects of cancer treatment that greatly reduce quality of life of cancer patients and survivors. Currently, there are no Food and Drug Administration-approved agents for the prevention or curative treatment of CIPN or CICI. The dual leucine zipper kinase (DLK) is a key mediator of axonal degeneration that is localized to axons and coordinates the neuronal response to injury. We developed a novel brain-penetrant DLK inhibitor, IACS'8287, which demonstrates potent and highly selective inhibition of DLK in vitro and in vivo. Coadministration of IACS'8287 with the platinum derivative cisplatin prevents mechanical allodynia, loss of intraepidermal nerve fibers in the hind paws, cognitive deficits, and impairments in brain connectivity in mice, all without interfering with the antitumor activity of cisplatin. The protective effects of IACS'8287 are associated with preservation of mitochondrial function in dorsal root ganglion neurons and in brain synaptosomes. In addition, RNA sequencing analysis of dorsal root ganglia reveals modulation of genes involved in neuronal activity and markers for immune cell infiltration by DLK inhibition. These data indicate that CIPN and CICI require DLK signaling in mice, and DLK inhibitors could become an attractive treatment in the clinic when coadministered with cisplatin, and potentially other chemotherapeutic agents, to prevent neurotoxicities as a result of cancer treatment.
Collapse
Affiliation(s)
- Jiacheng Ma
- The Neurodegeneration Consortium, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sunil Goodwani
- The Neurodegeneration Consortium, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Paul J. Acton
- The Neurodegeneration Consortium, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Virginie Buggia-Prevot
- The Neurodegeneration Consortium, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shelli R. Kesler
- Cancer Neuroscience Lab, School of Nursing, Department of Diagnostic Medicine, LIVESTRONG Cancer Institutes, University of Texas at Austin, Austin, TX, United States
| | - Imran Jamal
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Iteeben D. Mahant
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Zhen Liu
- Institute for Applied Cancer Science, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Faika Mseeh
- Institute for Applied Cancer Science, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Bruce L. Roth
- The Neurodegeneration Consortium, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Chaitali Chakraborty
- The Neurodegeneration Consortium, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Bo Peng
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Qi Wu
- Institute for Applied Cancer Science, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yongying Jiang
- Institute for Applied Cancer Science, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kang Le
- Institute for Applied Cancer Science, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael J. Soth
- Institute for Applied Cancer Science, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Philip Jones
- Institute for Applied Cancer Science, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - William J. Ray
- The Neurodegeneration Consortium, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cobi J. Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
34
|
Kasembeli MM, Singhmar P, Ma J, Edralin J, Tang Y, Adams C, Heijnen CJ, Kavelaars A, Tweardy DJ. TTI-101: A competitive inhibitor of STAT3 that spares oxidative phosphorylation and reverses mechanical allodynia in mouse models of neuropathic pain. Biochem Pharmacol 2021; 192:114688. [PMID: 34274354 PMCID: PMC8478865 DOI: 10.1016/j.bcp.2021.114688] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 01/06/2023]
Abstract
Signal Transducer and Activator of Transcription (STAT) 3 emerged rapidly as a high-value target for treatment of cancer. However, small-molecule STAT3 inhibitors have been slow to enter the clinic due, in part, to serious adverse events (SAE), including lactic acidosis and peripheral neuropathy, which have been attributed to inhibition of STAT3's mitochondrial function. Our group developed TTI-101, a competitive inhibitor of STAT3 that targets the receptor pY705-peptide binding site within the Src homology 2 (SH2) domain to block its recruitment and activation. TTI-101 has shown target engagement, no toxicity, and evidence of clinical benefit in a Phase I study in patients with solid tumors. Here we report that TTI-101 did not affect mitochondrial function, nor did it cause STAT3 aggregation, chemically modify STAT3 or cause neuropathic pain. Instead, TTI-101 unexpectedly suppressed neuropathic pain induced by chemotherapy or in a spared nerve injury model. Thus, in addition to its direct anti-tumor effect, TTI-101 may be of benefit when administered to cancer patients at risk of developing chemotherapy-induced peripheral neuropathy (CIPN).
Collapse
Affiliation(s)
- Moses M Kasembeli
- The Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Pooja Singhmar
- The Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Jiacheng Ma
- The Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Jules Edralin
- The Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Yongfu Tang
- The Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Clydell Adams
- The Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Cobi J Heijnen
- The Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Annemieke Kavelaars
- The Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - David J Tweardy
- The Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States.
| |
Collapse
|
35
|
Neuroprotective Potential of Bone Marrow-Derived Mesenchymal Stem Cells Following Chemotherapy. Biomedicines 2021; 9:biomedicines9070750. [PMID: 34209542 PMCID: PMC8301303 DOI: 10.3390/biomedicines9070750] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 11/17/2022] Open
Abstract
Cisplatin (CP) is extensively used in the medical oncology field for malignancy treatment, but its use is associated with neurological side effects that compromise the patients' quality of life. Cytotherapy is a new treatment strategy for tissue damage that has recently emerged. The use of bone marrow-derived mesenchymal stem cells (BM-MSCs) was investigated for its therapeutic potential against CP-induced chemobrain as well as various models of brain damage. This study was carried out to elucidate, for the first time, the role of the intravenous injection (IV) of BM-MSCs against CP-induced neurotoxicity in a rat model through investigation of the parameters of oxidative stress, inflammation, and apoptosis in brain tissue. A rat model of neurotoxicity was generated by intraperitoneal injection of 7.5 mg/kg CP while 2 × 106 BM-MSCs was given by IV as a therapeutic dose. Injection of CP led to a significant rise in malondialdehyde and nitric oxide levels accompanied by a marked depletion of superoxide dismutase and reduced glutathione content in brain tissue in comparison to the normal control (NC) rats. Furthermore, a remarkable rise in the brain levels of inflammatory cytokines interleukin (IL)-1β and IL-6, together with the expression of apoptotic marker caspase-3, and the downregulation of the brain expression of proliferating marker Ki-67 in brain tissue were detected in the CP group compared to the NC group. Histopathological alterations were observed in the brain tissue of the CP group. BM-MSCs mitigated the biochemical and histopathological alterations induced by CP without affecting brain cell proliferation. BM-MSCs could be used as a promising neuroprotective agent against CP-induced neurotoxicity.
Collapse
|
36
|
Harrison RA, Sharafeldin N, Rexer JL, Streck B, Petersen M, Henneghan AM, Kesler SR. Neurocognitive Impairment After Hematopoietic Stem Cell Transplant for Hematologic Malignancies: Phenotype and Mechanisms. Oncologist 2021; 26:e2021-e2033. [PMID: 34156729 DOI: 10.1002/onco.13867] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 06/14/2021] [Indexed: 12/16/2022] Open
Abstract
Hematopoietic stem cell transplant (HSCT) plays a central role in the treatment of hematologic cancers. With the increasing survival of patients after HSCT, survivorship issues experienced by this population have become an important outcome. Cognitive impairment is an established sequela of HSCT, with studies to date establishing its presence, associated risk factors, and clinical phenotype. There are multiple potential contributors to cognitive impairment after HSCT. Efforts are ongoing to further characterize its clinical phenotype, associated biomarkers, and biologic underpinnings. A fundamental knowledge of post-HSCT cognitive impairment is of value for all clinicians who interface with this population, and further academic efforts are needed to more fully understand the impact of this cancer treatment on brain health. IMPLICATIONS FOR PRACTICE: As survival outcomes after hematopoietic stem cell transplant (HSCT) improve, an awareness of the post-treatment challenges faced by this population has become central to its care. HSCT can have a sustained and broad impact on brain health, causing cognitive dysfunction, fatigue, disturbed mood, and sleep. In affected patients, autonomy, return to work, relationships, and quality of life may all be affected. A fundamental fluency in this area is important for clinicians interfacing with HSCT survivors, facilitating the identification and management of cognitive dysfunction and concurrent symptom clusters, and stimulating interest in these sequelae as areas for future clinical research.
Collapse
Affiliation(s)
- Rebecca A Harrison
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Noha Sharafeldin
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jennie L Rexer
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Brennan Streck
- Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Melissa Petersen
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Ashley M Henneghan
- School of Nursing, Dell School of Medicine, University of Texas at Austin, Austin, Texas, USA.,Department of Oncology, Dell School of Medicine, University of Texas at Austin, Austin, Texas, USA
| | - Shelli R Kesler
- School of Nursing, Dell School of Medicine, University of Texas at Austin, Austin, Texas, USA.,Department of Diagnostic Medicine, Dell School of Medicine, University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
37
|
El-Derany MO, Noureldein MH. Bone marrow mesenchymal stem cells and their derived exosomes resolve doxorubicin-induced chemobrain: critical role of their miRNA cargo. Stem Cell Res Ther 2021; 12:322. [PMID: 34090498 PMCID: PMC8180158 DOI: 10.1186/s13287-021-02384-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Background Doxorubicin (DOX), a widely used chemotherapeutic agent, can cause neurodegeneration in the brain, which leads to a condition known as chemobrain. In fact, chemobrain is a deteriorating condition which adversely affects the lives of cancer survivors. This study aimed to examine the potential therapeutic effects of bone marrow mesenchymal stem cells (BMSCs) and their derived exosomes (BMSCs-Exo) in DOX-induced chemobrain in rat models. Methods Chemobrain was induced by exposing rats to DOX (2 mg/kg, i.p) once weekly for 4 consecutive weeks. After 48 h of the last DOX dose, a subset of rats was supplied with either an intravenous injection of BMSCs (1 × 106) or a single dose of 150 μg of BMSCs-Exo. Behavioral tests were conducted 7 days post injection. Rats were sacrificed after 14 days from BMSCs or BMSCs-Exo injection. Results BMSCs and BMSCs-Exo successfully restored DOX-induced cognitive and behavioral distortion. These actions were mediated via decreasing hippocampal neurodegeneration and neural demyelination through upregulating neural myelination factors (myelin%, Olig2, Opalin expression), neurotropic growth factors (BDNF, FGF-2), synaptic factors (synaptophysin), and fractalkine receptor expression (Cx3cr1). Halting neurodegeneration in DOX-induced chemobrain was achieved through epigenetic induction of key factors in Wnt/β-catenin and hedgehog signaling pathways mediated primarily by the most abundant secreted exosomal miRNAs (miR-21-5p, miR-125b-5p, miR-199a-3p, miR-24-3p, let-7a-5p). Moreover, BMSCs and BMSCs-Exo significantly abrogate the inflammatory state (IL-6, TNF-α), apoptotic state (BAX/Bcl2), astrocyte, and microglia activation (GFAP, IBA-1) in DOX-induced chemobrain with a significant increase in the antioxidant mediators (GSH, GPx, SOD activity). Conclusions BMSCs and their derived exosomes offer neuroprotection against DOX-induced chemobrain via genetic and epigenetic abrogation of hippocampal neurodegeneration through modulating Wnt/β-catenin and hedgehog signaling pathways and through reducing inflammatory, apoptotic, and oxidative stress state. Graphical abstract Proposed mechanisms of the protective effects of bone marrow stem cells (BMSCs) and their exosomes (BMSCs-Exo) in doxorubicin (DOX)-induced chemobrain. Blue arrows: induce. Red arrows: inhibit.
![]() Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02384-9.
Collapse
Affiliation(s)
- Marwa O El-Derany
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
| | - Mohamed H Noureldein
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.,Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,American University of Beirut Diabetes Program, Beirut, Lebanon
| |
Collapse
|
38
|
Gibson EM, Monje M. Microglia in Cancer Therapy-Related Cognitive Impairment. Trends Neurosci 2021; 44:441-451. [PMID: 33674135 PMCID: PMC8593823 DOI: 10.1016/j.tins.2021.02.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 01/20/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022]
Abstract
Millions of cancer survivors experience a persistent neurological syndrome that includes deficits in memory, attention, information processing, and mental health. Cancer therapy-related cognitive impairment can cause mild to severe disruptions to quality of life for these cancer survivors. Understanding the cellular and molecular underpinnings of this disorder will facilitate new therapeutic strategies aimed at ameliorating these long-lasting impairments. Accumulating evidence suggests that a range of cancer therapies induce persistent activation of the brain's resident immune cells, microglia. Cancer therapy-induced microglial activation disrupts numerous mechanisms of neuroplasticity, and emerging findings suggest that this impairment in plasticity is central to cancer therapy-related cognitive impairment. This review explores reactive microglial dysregulation of neural circuit structure and function following cancer therapy.
Collapse
Affiliation(s)
- Erin M Gibson
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA 94305, USA.
| | - Michelle Monje
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA 94305, USA; Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA 94305, USA; Department of Pathology, Stanford University, Palo Alto, CA 94305, USA; Stanford California Department of Pediatrics, Stanford University, Palo Alto, CA 94305, USA.
| |
Collapse
|
39
|
Kesler SR, Sleurs C, McDonald BC, Deprez S, van der Plas E, Nieman BJ. Brain Imaging in Pediatric Cancer Survivors: Correlates of Cognitive Impairment. J Clin Oncol 2021; 39:1775-1785. [PMID: 33886371 DOI: 10.1200/jco.20.02315] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Shelli R Kesler
- School of Nursing, Department of Diagnostic Medicine, Dell School of Medicine, Livestrong Cancer Institutes, Austin, TX
| | - Charlotte Sleurs
- Department of Oncology, Catholic University of Leuven, Leuven, Belgium.,Leuven Cancer Institute, Leuven, Belgium
| | - Brenna C McDonald
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Center for Neuroimaging, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN
| | - Sabine Deprez
- Leuven Cancer Institute, Leuven, Belgium.,Department of Imaging and Pathology, Catholic University of Leuven, Leuven, Belgium
| | - Ellen van der Plas
- Department of Psychiatry, University of Iowa Hospital and Clinics, Iowa City, Iowa
| | - Brian J Nieman
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Ontario Institute for Cancer Research, Toronto, ON, Canada.,Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
40
|
Lomeli N, Lepe J, Gupta K, Bota DA. Cognitive complications of cancer and cancer-related treatments - Novel paradigms. Neurosci Lett 2021; 749:135720. [PMID: 33582187 PMCID: PMC8423125 DOI: 10.1016/j.neulet.2021.135720] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 01/07/2023]
Abstract
As advances in diagnostics and therapeutic strategies in oncology have increased the number of cancer survivors, the investigation of the mechanisms associated with long-term cognitive complications of cancer treatment has become an important topic of interest. The neurotoxic effects of chemotherapeutic agents have been described in pre-clinical and clinical research. In vitro and rodent studies have identified some underlying mechanisms contributing to chemotherapy-induced neurotoxicity and cognitive impairment for various chemotherapy drugs and other cancer treatments. However, investigation of the direct biological effects of cancer and other potential contributing factors in the pathogenesis of cancer-related cognitive impairment (CRCI) has only recently come into focus. This review will highlight evidence from pre-clinical tumor-bearing rodent models suggesting that cancer influences the cognitive and behavioral changes reported in human cancer populations through direct or indirect pathways that alter the normal neuroinflammatory responses, induce structural brain deficits, and decrease neurogenesis. We reflect on human clinical cancer research indicating that cognitive and behavioral changes precede cancer treatment in some malignancies. We also highlight implications for future areas of CRCI research based on novel findings on the interplay between cancer, chemotherapy, inflammation, tau pathology, and dysregulation of the microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Naomi Lomeli
- Department of Neurology, University of California Irvine, Irvine, CA, USA.
| | - Javier Lepe
- Department of Pathology & Laboratory Medicine, University of California Irvine, Irvine, CA, USA.
| | - Kalpna Gupta
- Department of Medicine, University of California Irvine, Irvine, CA, USA.
| | - Daniela A Bota
- Department of Neurology, University of California Irvine, Irvine, CA, USA; Department of Pathology & Laboratory Medicine, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
41
|
Boukelmoune N, Laumet G, Tang Y, Ma J, Mahant I, Nijboer C, Benders M, Kavelaars A, Heijnen CJ, Heijnen CJ. Nasal administration of mesenchymal stem cells reverses chemotherapy-induced peripheral neuropathy in mice. Brain Behav Immun 2021; 93:43-54. [PMID: 33316379 PMCID: PMC8826497 DOI: 10.1016/j.bbi.2020.12.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/18/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is one of the most frequently reported adverse effects of cancer treatment. CIPN often persists long after treatment completion and has detrimental effects on patient's quality of life. There are no efficacious FDA-approved drugs for CIPN. We recently demonstrated that nasal administration of mesenchymal stem cells (MSC) reverses the cognitive deficits induced by cisplatin in mice. Here we show that nasal administration of MSC after cisplatin- or paclitaxel treatment- completely reverses signs of established CIPN, including mechanical allodynia, spontaneous pain, and loss of intraepidermal nerve fibers (IENF) in the paw. The resolution of CIPN is associated with normalization of the cisplatin-induced decrease in mitochondrial bioenergetics in DRG neurons. Nasally administered MSC enter rapidly the meninges of the brain, spinal cord and peripheral lymph nodes to promote IL-10 production by macrophages. MSC-mediated resolution of mechanical allodynia, recovery of IENFs and restoration of DRG mitochondrial function critically depends on IL-10 production. MSC from IL-10 knockout animals are not capable of reversing the symptoms of CIPN. Moreover, WT MSC do not reverse CIPN in mice lacking IL-10 receptors on peripheral sensory neurons. In conclusion, only two nasal administrations of MSC fully reverse CIPN and the associated mitochondrial abnormalities via an IL-10 dependent pathway. Since MSC are already applied clinically, we propose that nasal MSC treatment could become a powerful treatment for the large group of patients suffering from neurotoxicities of cancer treatment.
Collapse
Affiliation(s)
- Nabila Boukelmoune
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA
| | - Geoffroy Laumet
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA.,Current affiliation: Department of Physiology, Michigan State University, East Lansing, Michigan, 48824, USA
| | - Yongfu Tang
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA
| | - Jiacheng Ma
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA
| | - Itee Mahant
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA
| | - Cora Nijboer
- Department of Developmental Origins of Disease, Division Woman and Baby, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Manon Benders
- Department of Neonatology, Division Woman and Baby, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA
| | - Cobi J. Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA.,Corresponding author at: Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Z8.5034, Houston, Texas, 77030. (Cobi J. Heijnen)
| | - Cobi J Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, TX 77030, USA.
| |
Collapse
|
42
|
Klontzas ME, Kakkos GA, Papadakis GZ, Marias K, Karantanas AH. Advanced clinical imaging for the evaluation of stem cell based therapies. Expert Opin Biol Ther 2021; 21:1253-1264. [PMID: 33576278 DOI: 10.1080/14712598.2021.1890711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: As stem cell treatments reach closer to the clinic, the need for appropriate noninvasive imaging for accurate disease diagnosis, treatment planning, follow-up, and early detection of complications, is constantly rising. Clinical radiology affords an extensive arsenal of advanced imaging techniques, to provide anatomical and functional information on the whole spectrum of stem cell treatments from diagnosis to follow-up.Areas covered: This manuscript aims at providing a critical review of major published studies on the utilization of advanced imaging for stem cell treatments. Uses of magnetic resonance imaging (MRI), computed tomography (CT), ultrasound, and positron emission tomography (PET) are reviewed and interrogated for their applicability to stem cell imaging.Expert opinion: A wide spectrum of imaging methods have been utilized for the evaluation of stem cell therapies. The majority of published techniques are not clinically applicable, using methods exclusively applicable to animals or technology irrelevant to current clinical practice. Harmonization of preclinical methods with clinical reality is necessary for the timely translation of stem cell therapies to the clinic. Methods such as diffusion weighted MRI, hybrid imaging, and contrast-enhanced ultrasound hold great promise and should be routinely incorporated in the evaluation of patients receiving stem cell treatments.
Collapse
Affiliation(s)
- Michail E Klontzas
- Department of Medical Imaging, University Hospital of Heraklion, Crete, Greece.,Advanced Hybrid Imaging Systems, Institute of Computer Science, Foundation for Research and Technology (FORTH), Heraklion, Crete, Greece
| | - George A Kakkos
- Department of Medical Imaging, University Hospital of Heraklion, Crete, Greece
| | - Georgios Z Papadakis
- Advanced Hybrid Imaging Systems, Institute of Computer Science, Foundation for Research and Technology (FORTH), Heraklion, Crete, Greece.,Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), Heraklion, Crete, Greece.,Department of Radiology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Kostas Marias
- Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), Heraklion, Crete, Greece.,Department of Electrical and Computer Engineering, Hellenic Mediterranean University, Heraklion, Crete, Greece
| | - Apostolos H Karantanas
- Department of Medical Imaging, University Hospital of Heraklion, Crete, Greece.,Advanced Hybrid Imaging Systems, Institute of Computer Science, Foundation for Research and Technology (FORTH), Heraklion, Crete, Greece.,Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), Heraklion, Crete, Greece.,Department of Radiology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| |
Collapse
|
43
|
Alexander JF, Seua AV, Arroyo LD, Ray PR, Wangzhou A, Heiβ-Lückemann L, Schedlowski M, Price TJ, Kavelaars A, Heijnen CJ. Nasal administration of mitochondria reverses chemotherapy-induced cognitive deficits. Theranostics 2021; 11:3109-3130. [PMID: 33537077 PMCID: PMC7847685 DOI: 10.7150/thno.53474] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 12/12/2020] [Indexed: 12/15/2022] Open
Abstract
Up to seventy-five percent of patients treated for cancer suffer from cognitive deficits which can persist for months to decades, severely impairing quality of life. Although the number of cancer survivors is increasing tremendously, no efficacious interventions exist. Cisplatin, most commonly employed for solid tumors, leads to cognitive impairment including deficits in memory and executive functioning. We recently proposed deficient neuronal mitochondrial function as its underlying mechanism. We hypothesized nasal administration of mitochondria isolated from human mesenchymal stem cells to mice, can reverse cisplatin-induced cognitive deficits. Methods: Puzzle box, novel object place recognition and Y-maze tests were used to assess the cognitive function of mice. Immunofluorescence and high-resolution confocal microscopy were employed to trace the nasally delivered mitochondria and evaluate their effect on synaptic loss. Black Gold II immunostaining was used to determine myelin integrity. Transmission electron microscopy helped determine mitochondrial and membrane integrity of brain synaptosomes. RNA-sequencing was performed to analyse the hippocampal transcriptome. Results: Two nasal administrations of mitochondria isolated from human mesenchymal stem cells to mice, restored executive functioning, working and spatial memory. Confocal imaging revealed nasally delivered mitochondria rapidly arrived in the meninges where they were readily internalized by macrophages. The administered mitochondria also accessed the rostral migratory stream and various other brain regions including the hippocampus where they colocalized with GFAP+ cells. The restoration of cognitive function was associated with structural repair of myelin in the cingulate cortex and synaptic loss in the hippocampus. Nasal mitochondrial donation also reversed the underlying synaptosomal mitochondrial defects. Moreover, transcriptome analysis by RNA-sequencing showed reversal of cisplatin-induced changes in the expression of about seven hundred genes in the hippocampus. Pathway analysis identified Nrf2-mediated response as the top canonical pathway. Conclusion: Our results provide key evidence on the therapeutic potential of isolated mitochondria - restoring both brain structure and function, their capability to enter brain meninges and parenchyma upon nasal delivery and undergo rapid cellular internalization and alter the hippocampal transcriptome. Our data identify nasal administration of mitochondria as an effective strategy for reversing chemotherapy-induced cognitive deficits and restoring brain health, providing promise for the growing population of both adult and pediatric cancer survivors.
Collapse
Affiliation(s)
- Jenolyn F. Alexander
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, United States
| | - Alexandre V. Seua
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, United States
| | - Luis D. Arroyo
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, United States
| | - Pradipta R. Ray
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, Texas, United States
| | - Andi Wangzhou
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, Texas, United States
| | - Laura Heiβ-Lückemann
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Theodore J. Price
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, Texas, United States
| | - Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, United States
| | - Cobi J. Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, United States
| |
Collapse
|
44
|
Fathollahi A, Hashemi SM, Haji Molla Hoseini M, Tavakoli S, Farahani E, Yeganeh F. Intranasal administration of small extracellular vesicles derived from mesenchymal stem cells ameliorated the experimental autoimmune encephalomyelitis. Int Immunopharmacol 2021; 90:107207. [PMID: 33290966 DOI: 10.1016/j.intimp.2020.107207] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/26/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a mouse model for the human multiple sclerosis, which is characterized by inflammation in the central nervous system (CNS), de-myelination of axonal neurons, and loss of motor coordination. The aim of the current study was to evaluate the effect of intranasal administration of mesenchymal stem cells (MSCs) and small extracellular vesicle (SEV) derived from the MSC (MSC-SEV) on disease activity and antigen-specific responses in the EAE mouse model. MSCs (5 × 105) were administered intranasally to EAE mice (n = 5) on the 15th and 24th days after immunization. In addition, the intranasal administration of MSC-SEV (10 μg) was used to treat EAE mice (n = 5) on a daily basis from the 15th to the 27th day after induction of the disease. The outcomes of therapies were evaluated using studying clinical symptoms and histological analysis of CNS lesions. Moreover, T cell proliferation, the frequency of regulatory T cells, the expression of transcription factors of T-helper subsets, and the levels of their corresponded cytokines were evaluated in splenocytes culture that was stimulated with specific-antigen. The results of treatment of EAE mice with MSC- SEV and MSC showed a significant decrease in the clinical scores, and it was found that treatment with MSC-SEV was more effective in alleviating clinical scores than MSC. In addition, the decrease in clinical symptoms was associated with an increase in immunomodulatory responses, including an increase in the frequency of Foxp3+ CD25+ regulatory T cells. Moreover, the level of TGF-β was increased by both treatments; however, interleukin-10 was increased only by MSC treatment. Ultimately, it was achieved that the intranasal administration of MSC-SEV to EAE mice was more effective than the administration of MSC to reduce clinical scores and histological lesions of the CNS tissue.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Cytokines/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/surgery
- Extracellular Vesicles/immunology
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/transplantation
- Female
- Gene Expression Regulation
- Inflammation Mediators/metabolism
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/immunology
- Mesenchymal Stem Cells/metabolism
- Mice, Inbred C57BL
- Spinal Cord/immunology
- Spinal Cord/metabolism
- Spinal Cord/pathology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Mice
Collapse
Affiliation(s)
- Anwar Fathollahi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Haji Molla Hoseini
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajjad Tavakoli
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Elnaz Farahani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Farshid Yeganeh
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
Chiang ACA, Seua AV, Singhmar P, Arroyo LD, Mahalingam R, Hu J, Kavelaars A, Heijnen CJ. Bexarotene normalizes chemotherapy-induced myelin decompaction and reverses cognitive and sensorimotor deficits in mice. Acta Neuropathol Commun 2020; 8:193. [PMID: 33183353 PMCID: PMC7661216 DOI: 10.1186/s40478-020-01061-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/17/2020] [Indexed: 12/20/2022] Open
Abstract
Frequently reported neurotoxic sequelae of cancer treatment include cognitive deficits and sensorimotor abnormalities that have long-lasting negative effects on the quality of life of an increasing number of cancer survivors. The underlying mechanisms are not fully understood and there is no effective treatment. We show here that cisplatin treatment of mice not only caused cognitive dysfunction but also impaired sensorimotor function. These functional deficits are associated with reduced myelin density and complexity in the cingulate and sensorimotor cortex. At the ultrastructural level, myelin abnormalities were characterized by decompaction. We used this model to examine the effect of bexarotene, an agonist of the RXR-family of nuclear receptors. Administration of only five daily doses of bexarotene after completion of cisplatin treatment was sufficient to normalize myelin density and fiber coherency and to restore myelin compaction in cingulate and sensorimotor cortex. Functionally, bexarotene normalized performance of cisplatin-treated mice in tests for cognitive and sensorimotor function. RNAseq analysis identified the TR/RXR pathway as one of the top canonical pathways activated by administration of bexarotene to cisplatin-treated mice. Bexarotene also activated neuregulin and netrin pathways that are implicated in myelin formation/maintenance, synaptic function and axonal guidance. In conclusion, short term treatment with bexarotene is sufficient to reverse the adverse effects of cisplatin on white matter structure, cognitive function, and sensorimotor performance. These encouraging findings warrant further studies into potential clinical translation and the underlying mechanisms of bexarotene for chemobrain.
Collapse
|
46
|
Cell-specific role of histone deacetylase 6 in chemotherapy-induced mechanical allodynia and loss of intraepidermal nerve fibers. Pain 2020; 160:2877-2890. [PMID: 31356453 DOI: 10.1097/j.pain.0000000000001667] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a serious adverse side effect of cancer treatment with no Food and Drug Administration-approved medication for its prevention or management. Using RNA sequencing analysis of dorsal root ganglia (DRG), we identify critical contributions of histone deacetylase 6 (HDAC6) and mitochondrial damage to the establishment of CIPN in a mouse model of cisplatin-induced neuropathy. We show that pharmacological inhibition of HDAC6 using ACY-1215 or global deletion of HDAC6 is sufficient to prevent cisplatin-induced mechanical allodynia, loss of intraepidermal nerve fibers (IENFs), and mitochondrial bioenergetic deficits in DRG neurons and peripheral nerves in male and female mice. The bioenergetic deficits in the neuronal cell bodies in the DRG are characterized by reduced oxidative phosphorylation, whereas the mitochondrial deficits in the nerves are due to a reduction in axonal mitochondrial content. Notably, deleting HDAC6 in sensory neurons protects against the cisplatin-induced loss of IENFs and the reduction in mitochondrial bioenergetics and content in the peripheral nerve. By contrast, deletion of HDAC6 in sensory neurons only partially and transiently prevents cisplatin-induced mechanical allodynia and does not protect against impairment of mitochondrial function in DRG neurons. We further reveal a critical role of T cells in the protective effects of HDAC6 inhibition on these signs of CIPN. In summary, we show that cisplatin-induced mechanical allodynia is associated with mitochondrial damage in DRG neurons, whereas the loss of IENFs is related to bioenergetic deficits in peripheral nerves. Moreover, our findings identify cell-specific contributions of HDAC6 to mechanical allodynia and loss of IENFs that characterize cisplatin-induced peripheral neuropathy.
Collapse
|
47
|
Srivastava RK, Singh P. Stem cell therapies as a therapeutic option to counter chemo brain: a negative effect of cancer treatment. Regen Med 2020; 15:1789-1800. [PMID: 32844724 DOI: 10.2217/rme-2020-0060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Chemo brain, a constellation of cognitive deficiencies followed by chemotherapy drugs, used to treat different types of cancers and adversely impacts the quality of life of a cancer survivor. The underlying mechanism of chemo brain remains vague, thus delaying the advancement of efficient treatments. Unfortunately, there is no US FDA approved medicine for chemo brain and often medicines considered for chemo brain are already the ones approved for other diseases. Nevertheless, researches exploring stem cell transplantation in different neurodegenerative diseases demonstrate that cellular transplantation could reverse chemotherapy-induced chemo brain. This review talks about the mechanism behind the cognitive impairments instigated by different chemotherapy drugs used in cancer treatment, and how stem cell therapy could be advantageous to overcome this disease.
Collapse
Affiliation(s)
- Rohit K Srivastava
- Department of Pediatrics Surgery, Texas Children's Hospital, Houston, TX 77030, USA.,M.E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pratibha Singh
- Department of Biochemistry and Cell Biology, Biosciences Research Collaborative, Rice University, Houston, TX 77030, USA
| |
Collapse
|
48
|
Kesler SR, Petersen ML, Rao V, Harrison RA, Palesh O. Functional connectome biotypes of chemotherapy-related cognitive impairment. J Cancer Surviv 2020; 14:483-493. [PMID: 32157609 PMCID: PMC7958311 DOI: 10.1007/s11764-020-00863-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/31/2020] [Indexed: 12/31/2022]
Abstract
PURPOSE Cancer-related cognitive impairment (CRCI) is a common neurotoxicity among patients with breast and other cancers. Neuroimaging studies have demonstrated measurable biomarkers of CRCI but have largely neglected the potential heterogeneity of the syndrome. METHODS We used retrospective functional MRI data from 80 chemotherapy-treated breast cancer survivors to examine neurophysiologic subtypes or "biotypes" of CRCI. The breast cancer group consisted of training (N = 57) and validation (N = 23) samples. RESULTS An unsupervised clustering approach using connectomes from the training sample identified three distinct biotypes. Cognitive performance (p < 0.05, corrected) and regional connectome organization (p < 0.001, corrected) differed significantly between the biotypes and also from 103 healthy female controls. We then built a random forest classifier using connectome features to distinguish between the biotypes (accuracy = 91%) and applied this to the validation sample to predict biotype assignment. Cognitive performance (p < 0.05, corrected) and regional connectome organization (p < 0.005, corrected) differed significantly between the predicted biotypes and healthy controls. Biotypes were also characterized by divergent clinical and demographic factors as well as patient reported outcomes. CONCLUSIONS Neurophysiologic biotypes may help characterize the heterogeneity associated with CRCI in a data-driven manner based on neuroimaging biomarkers. IMPLICATIONS FOR CANCER SURVIVORS Our novel findings provide a foundation for detecting potential risk and resilience factors that warrant further study. With further investigation, biotypes might be used to personalize assessments of and interventions for CRCI.
Collapse
Affiliation(s)
- Shelli R Kesler
- Cancer Neuroscience Laboratory, School of Nursing, University of Texas at Austin, 1710 Red River St, Austin, TX, 78712, USA.
- Department of Diagnostic Medicine, Dell School of Medicine, University of Texas at Austin, Austin, TX, USA.
- LIVESTRONG Cancer Institutes, Dell School of Medicine, University of Texas at Austin, Austin, TX, USA.
| | - Melissa L Petersen
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Vikram Rao
- Cancer Neuroscience Laboratory, School of Nursing, University of Texas at Austin, 1710 Red River St, Austin, TX, 78712, USA
- Department of Diagnostic Medicine, Dell School of Medicine, University of Texas at Austin, Austin, TX, USA
| | - Rebecca A Harrison
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Oxana Palesh
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
- Stanford Cancer Institute, Palo Alto, CA, USA
| |
Collapse
|
49
|
Stavely R, Nurgali K. The emerging antioxidant paradigm of mesenchymal stem cell therapy. Stem Cells Transl Med 2020; 9:985-1006. [PMID: 32497410 PMCID: PMC7445024 DOI: 10.1002/sctm.19-0446] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/05/2020] [Accepted: 04/20/2020] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (multipotent stromal cells; MSCs) have been under investigation for the treatment of diverse diseases, with many promising outcomes achieved in animal models and clinical trials. The biological activity of MSC therapies has not been fully resolved which is critical to rationalizing their use and developing strategies to enhance treatment efficacy. Different paradigms have been constructed to explain their mechanism of action, including tissue regeneration, trophic/anti-inflammatory secretion, and immunomodulation. MSCs rarely engraft and differentiate into other cell types after in vivo administration. Furthermore, it is equivocal whether MSCs function via the secretion of many peptide/protein ligands as their therapeutic properties are observed across xenogeneic barriers, which is suggestive of mechanisms involving mediators conserved between species. Oxidative stress is concomitant with cellular injury, inflammation, and dysregulated metabolism which are involved in many pathologies. Growing evidence supports that MSCs exert antioxidant properties in a variety of animal models of disease, which may explain their cytoprotective and anti-inflammatory properties. In this review, evidence of the antioxidant effects of MSCs in in vivo and in vitro models is explored and potential mechanisms of these effects are discussed. These include direct scavenging of free radicals, promoting endogenous antioxidant defenses, immunomodulation via reactive oxygen species suppression, altering mitochondrial bioenergetics, and donating functional mitochondria to damaged cells. Modulation of the redox environment and oxidative stress by MSCs can mediate their anti-inflammatory and cytoprotective properties and may offer an explanation to the diversity in disease models treatable by MSCs and how these mechanisms may be conserved between species.
Collapse
Affiliation(s)
- Rhian Stavely
- Institute for Health and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia.,Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia.,Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia.,Regenerative Medicine and Stem Cells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, Victoria, Australia
| |
Collapse
|
50
|
English K, Shepherd A, Uzor NE, Trinh R, Kavelaars A, Heijnen CJ. Astrocytes rescue neuronal health after cisplatin treatment through mitochondrial transfer. Acta Neuropathol Commun 2020; 8:36. [PMID: 32197663 PMCID: PMC7082981 DOI: 10.1186/s40478-020-00897-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/13/2020] [Indexed: 12/28/2022] Open
Abstract
Neurodegenerative disorders, including chemotherapy-induced cognitive impairment, are associated with neuronal mitochondrial dysfunction. Cisplatin, a commonly used chemotherapeutic, induces neuronal mitochondrial dysfunction in vivo and in vitro. Astrocytes are key players in supporting neuronal development, synaptogenesis, axonal growth, metabolism and, potentially mitochondrial health. We tested the hypothesis that astrocytes transfer healthy mitochondria to neurons after cisplatin treatment to restore neuronal health.We used an in vitro system in which astrocytes containing mito-mCherry-labeled mitochondria were co-cultured with primary cortical neurons damaged by cisplatin. Culture of primary cortical neurons with cisplatin reduced neuronal survival and depolarized neuronal mitochondrial membrane potential. Cisplatin induced abnormalities in neuronal calcium dynamics that were characterized by increased resting calcium levels, reduced calcium responses to stimulation with KCl, and slower calcium clearance. The same dose of cisplatin that caused neuronal damage did not affect astrocyte survival or astrocytic mitochondrial respiration. Co-culture of cisplatin-treated neurons with astrocytes increased neuronal survival, restored neuronal mitochondrial membrane potential, and normalized neuronal calcium dynamics especially in neurons that had received mitochondria from astrocytes which underlines the importance of mitochondrial transfer. These beneficial effects of astrocytes were associated with transfer of mitochondria from astrocytes to cisplatin-treated neurons. We show that siRNA-mediated knockdown of the Rho-GTPase Miro-1 in astrocytes reduced mitochondrial transfer from astrocytes to neurons and prevented the normalization of neuronal calcium dynamics.In conclusion, we showed that transfer of mitochondria from astrocytes to neurons rescues neurons from the damage induced by cisplatin treatment. Astrocytes are far more resistant to cisplatin than cortical neurons. We propose that transfer of functional mitochondria from astrocytes to neurons is an important repair mechanism to protect the vulnerable cortical neurons against the toxic effects of cisplatin.
Collapse
Affiliation(s)
- Krystal English
- Division of Internal Medicine, Department of Symptom Research, Laboratories of Neuroimmunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Andrew Shepherd
- Division of Internal Medicine, Department of Symptom Research, Laboratories of Neuroimmunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Ndidi-Ese Uzor
- Department of Neurobiology & Anatomy, The University of Texas McGovern Medical School, Houston, TX 77030 USA
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX 77030 USA
| | - Ronnie Trinh
- Division of Internal Medicine, Department of Symptom Research, Laboratories of Neuroimmunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Annemieke Kavelaars
- Division of Internal Medicine, Department of Symptom Research, Laboratories of Neuroimmunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Cobi J. Heijnen
- Division of Internal Medicine, Department of Symptom Research, Laboratories of Neuroimmunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| |
Collapse
|