1
|
Mortensen C, Thomsen MT, Chua KC, Hammer HS, Nielsen F, Pötz O, Svenningsen AF, Kroetz DL, Stage TB. Modeling mechanisms of chemotherapy-induced peripheral neuropathy and chemotherapy transport using induced pluripotent stem cell-derived sensory neurons. Neuropharmacology 2024; 258:110062. [PMID: 38972371 DOI: 10.1016/j.neuropharm.2024.110062] [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: 02/19/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND and Purpose: Chemotherapy-induced peripheral neuropathy (CIPN) constitutes a significant health problem due to the increasing prevalence and lack of therapies for treatment and prevention. While pivotal for routine cancer treatment, paclitaxel and vincristine frequently cause CIPN and impact the quality of life among cancer patients and survivors. Here, we investigate molecular mechanisms and drug transport in CIPN. EXPERIMENTAL APPROACH Human sensory neurons were derived from induced pluripotent stem cells (iPSC-SNs), which were characterized using flow cytometry and immunolabeling. These iPSC-SNs were exposed to different concentrations of the two microtubule-targeting agents, paclitaxel and vincristine, with and without pre-exposure to inhibitors and inducers of efflux transporters. Neuronal networks were quantified via fluorescent staining against sensory neuron markers. Transcriptional effects of the chemotherapeutics were examined using quantitative polymerase chain reactions (qPCR). KEY RESULTS Paclitaxel exposure resulted in axonal retraction and thickening, while vincristine caused fragmentation and abolishment of axons. Both agents increased the mRNA expression of the pain receptor, transient receptor potential vanilloid (TRPV1), and highly induced neuronal damage, as measured by activating transcription factor 3 (ATF3) mRNA. iPSC-SNs express the efflux transporters, P-glycoprotein (P-gp, encoded by ABCB1) and multidrug resistance-associated protein 1 (MPR1, encoded by ABCC1). Modulation of efflux transporters indicate that P-gp and MRP1 play a role in modulating neuronal accumulation and neurotoxicity in preliminary experiments. CONCLUSION and Implications: iPSC-SNs are a valuable and robust model to study the role of efflux transporters and other mechanistic targets in CIPN. Efflux transporters may play a role in CIPN pathogenesis as they regulate the disposition of chemotherapy to the peripheral nervous system, and they may present potential therapeutic targets for CIPN.
Collapse
Affiliation(s)
- Christina Mortensen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Mikkel Thy Thomsen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Katherina C Chua
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | | | - Flemming Nielsen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | | | - Asa Fex Svenningsen
- Neurobiology Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Deanna L Kroetz
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Tore Bjerregaard Stage
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark; Department of Clinical Pharmacology, Odense University Hospital, Odense, Denmark.
| |
Collapse
|
2
|
Tarasiuk O, Invernizzi C, Alberti P. In vitro neurotoxicity testing: lessons from chemotherapy-induced peripheral neurotoxicity. Expert Opin Drug Metab Toxicol 2024:1-16. [PMID: 39246127 DOI: 10.1080/17425255.2024.2401584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION Chemotherapy induced peripheral neurotoxicity (CIPN) is a long-lasting, or even permanent, late toxicity caused by largely used anticancer drugs. CIPN affects a growing population of cancer survivors and diminishes their quality of life since there is no curative/preventive treatment. Among several reasons for this unmet clinical need, there is an incomplete knowledge on mechanisms leading to CIPN. Therefore, bench side research is still greatly needed: in vitro studies are pivotal to both evaluate neurotoxicity mechanisms and potential neuroprotection strategies. AREAS COVERED Advantages and disadvantages of in vitro approaches are addressed with respect to their applicability to the CIPN field. Different cell cultures and techniques to assess neurotoxicity/neuroprotection are described. PubMed search-string: (chemotherapy-induced) AND (((neuropathy) OR neurotoxicity) OR neuropathic pain) AND (in vitro) AND (((((model) OR SH-SY5Y) OR PC12) OR iPSC) OR DRG neurons); (chemotherapy-induced) AND (((neuropathy) OR neurotoxicity) OR neuropathic pain) AND (model) AND (((neurite elongation) OR cell viability) OR morphology). No articles published before 1990 were selected. EXPERT OPINION CIPN is an ideal experimental setting to test axonal damage and, in general, peripheral nervous system mechanisms of disease and neuroprotection. Therefore, starting from robust preclinical data in this field, potentially, relevant biological rationale can be transferred to other human spontaneous diseases of the peripheral nervous system.
Collapse
Affiliation(s)
- Olga Tarasiuk
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- NeuroMI (Milan Center for Neuroscience), Milan, Italy
| | - Chiara Invernizzi
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- Neuroscience, School of Medicine and Surgery, Monza, Italy
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- NeuroMI (Milan Center for Neuroscience), Milan, Italy
- Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
3
|
Smulders PS, Heikamp K, Hermanides J, Hollmann MW, ten Hoope W, Weber NC. Chemotherapy-induced peripheral neuropathy models constructed from human induced pluripotent stem cells and directly converted cells: a systematic review. Pain 2024; 165:1914-1925. [PMID: 38381959 PMCID: PMC11331829 DOI: 10.1097/j.pain.0000000000003193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 02/23/2024]
Abstract
ABSTRACT Developments in human cellular reprogramming now allow for the generation of human neurons for in vitro disease modelling. This technique has since been used for chemotherapy-induced peripheral neuropathy (CIPN) research, resulting in the description of numerous CIPN models constructed from human neurons. This systematic review provides a critical analysis of available models and their methodological considerations (ie, used cell type and source, CIPN induction strategy, and validation method) for prospective researchers aiming to incorporate human in vitro models of CIPN in their research. The search strategy was developed with assistance from a clinical librarian and conducted in MEDLINE (PubMed) and Embase (Ovid) on September 26, 2023. Twenty-six peer-reviewed experimental studies presenting original data about human reprogrammed nonmotor neuron cell culture systems and relevant market available chemotherapeutics drugs were included. Virtually, all recent reports modeled CIPN using nociceptive dorsal root ganglion neurons. Drugs known to cause the highest incidence of CIPN were most used. Furthermore, treatment effects were almost exclusively validated by the acute effects of chemotherapeutics on neurite dynamics and cytotoxicity parameters, enabling the extrapolation of the half-maximal inhibitory concentration for the 4 most used chemotherapeutics. Overall, substantial heterogeneity was observed in the way studies applied chemotherapy and reported their findings. We therefore propose 6 suggestions to improve the clinical relevance and appropriateness of human cellular reprogramming-derived CIPN models.
Collapse
Affiliation(s)
- Pascal S.H. Smulders
- Department of Anesthesiology, Amsterdam UMC location University of Amsterdam, Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Kim Heikamp
- Department of Anesthesiology, Amsterdam UMC location University of Amsterdam, Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Jeroen Hermanides
- Department of Anesthesiology, Amsterdam UMC location University of Amsterdam, Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Markus W. Hollmann
- Department of Anesthesiology, Amsterdam UMC location University of Amsterdam, Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Werner ten Hoope
- Department of Anesthesiology, Amsterdam UMC location University of Amsterdam, Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
- Department of Anesthesiology, Rijnstate Hospital, Arnhem, the Netherlands
| | - Nina C. Weber
- Department of Anesthesiology, Amsterdam UMC location University of Amsterdam, Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| |
Collapse
|
4
|
LeBlang CJ, Pazyra-Murphy MF, Silagi ES, Dasgupta S, Tsolias M, Miller T, Petrova V, Zhen S, Jovanovic V, Castellano D, Gerrish K, Ormanoglu P, Tristan C, Singeç I, Woolf CJ, Tasdemir-Yilmaz O, Segal RA. Satellite glial contact enhances differentiation and maturation of human iPSC-derived sensory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604966. [PMID: 39211268 PMCID: PMC11361066 DOI: 10.1101/2024.07.24.604966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Sensory neurons generated from induced pluripotent stem cells (iSNs) are used to model human peripheral neuropathies, however current differentiation protocols produce sensory neurons with an embryonic phenotype. Peripheral glial cells contact sensory neurons early in development and contribute to formation of the canonical pseudounipolar morphology, but these signals are not encompassed in current iSN differentiation protocols. Here, we show that terminal differentiation of iSNs in co-culture with rodent Dorsal Root Ganglion satellite glia (rSG) advances their differentiation and maturation. Co-cultured iSNs develop a pseudounipolar morphology through contact with rSGs. This transition depends on semaphorin-plexin guidance cues and on glial gap junction signaling. In addition to morphological changes, iSNs terminally differentiated in co-culture exhibit enhanced spontaneous action potential firing, more mature gene expression, and increased susceptibility to paclitaxel induced axonal degeneration. Thus, iSNs differentiated in coculture with rSGs provide a better model for investigating human peripheral neuropathies.
Collapse
|
5
|
Cantor EL, Shen F, Jiang G, Philips S, Schneider BP. Optimization of a human induced pluripotent stem cell-derived sensory neuron model for the in vitro evaluation of taxane-induced neurotoxicity. Sci Rep 2024; 14:19075. [PMID: 39154055 PMCID: PMC11330481 DOI: 10.1038/s41598-024-69280-z] [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: 03/13/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024] Open
Abstract
Human induced pluripotent stem cell-derived sensory neuron (iPSC-dSN) models are a valuable resource for the study of neurotoxicity but are affected by poor replicability and reproducibility, often due to a lack of optimization. Here, we identify experimental factors related to culture conditions that substantially impact cellular drug response in vitro and determine optimal conditions for improved replicability and reproducibility. Treatment duration and cell seeding density were both found to be significant factors, while cell line differences also contributed to variation. A replicable dose-response in viability was demonstrated after 48-h exposure to docetaxel or paclitaxel. Additionally, a replicable dose-dependent reduction in neurite outgrowth was demonstrated, demonstrating the applicability of the model for the examination of additional phenotypes. Overall, we have established an optimized iPSC-dSN model for the study of taxane-induced neurotoxicity.
Collapse
Affiliation(s)
- Erica L Cantor
- Hematology/Oncology Division, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fei Shen
- Hematology/Oncology Division, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Guanglong Jiang
- Medical and Molecular Genetics Division, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Santosh Philips
- Hematology/Oncology Division, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bryan P Schneider
- Hematology/Oncology Division, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
6
|
Petrova V, Snavely AR, Splaine J, Zhen S, Singh B, Pandey R, Chen K, Cheng A, Hermawan C, Barrett LB, Smith JA, Woolf CJ. Identification of novel neuroprotectants against vincristine-induced neurotoxicity in iPSC-derived neurons. Cell Mol Life Sci 2024; 81:315. [PMID: 39066803 PMCID: PMC11335239 DOI: 10.1007/s00018-024-05340-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/05/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a disabling side effect of cancer chemotherapy that can often limit treatment options for cancer patients or have life-long neurodegenerative consequences that reduce the patient's quality of life. CIPN is caused by the detrimental actions of various chemotherapeutic agents on peripheral axons. Currently, there are no approved preventative measures or treatment options for CIPN, highlighting the need for the discovery of novel therapeutics and improving our understanding of disease mechanisms. In this study, we utilized human-induced pluripotent stem cell (hiPSC)-derived motor neurons as a platform to mimic axonal damage after treatment with vincristine, a chemotherapeutic used for the treatment of breast cancers, osteosarcomas, and leukemia. We screened a total of 1902 small molecules for neuroprotective properties in rescuing vincristine-induced axon growth deficits. From our primary screen, we identified 38 hit compounds that were subjected to secondary dose response screens. Six compounds showed favorable pharmacological profiles - AZD7762, A-674563, Blebbistatin, Glesatinib, KW-2449, and Pelitinib, all novel neuroprotectants against vincristine toxicity to neurons. In addition, four of these six compounds also showed efficacy against vincristine-induced growth arrest in human iPSC-derived sensory neurons. In this study, we utilized high-throughput screening of a large library of compounds in a therapeutically relevant assay. We identified several novel compounds that are efficacious in protecting different neuronal subtypes from the toxicity induced by a common chemotherapeutic agent, vincristine which could have therapeutic potential in the clinic.
Collapse
Affiliation(s)
- Veselina Petrova
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrew R Snavely
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jennifer Splaine
- ICCB-Longwood Screening Facility, Harvard Medical School, 250 Longwood Avenue, Boston, MA, 02115, USA
| | - Shannon Zhen
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Bhagat Singh
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Roshan Pandey
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Kuchuan Chen
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Anya Cheng
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Crystal Hermawan
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Lee B Barrett
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jennifer A Smith
- ICCB-Longwood Screening Facility, Harvard Medical School, 250 Longwood Avenue, Boston, MA, 02115, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA.
- F.M. Kirby Neurobiology Center, Center for Life Science, 3 Blackfan Circle, Boston, MA, 02115, USA.
| |
Collapse
|
7
|
Petrova V, Snavely AR, Splaine J, Zhen S, Singh B, Pandey R, Chen K, Cheng A, Hermawan C, Barrett LB, Smith JA, Woolf C. Identification of novel neuroprotectants against vincristine-induced neurotoxicity in iPSC-derived neurons. RESEARCH SQUARE 2024:rs.3.rs-4545853. [PMID: 39011110 PMCID: PMC11247920 DOI: 10.21203/rs.3.rs-4545853/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a disabling side effect of cancer chemotherapy that can often limit treatment options for cancer patients or have life-long neurodegenerative consequences that reduce the patient's quality of life. CIPN is caused by the detrimental actions of various chemotherapeutic agents on peripheral axons. Currently, there are no approved preventative measures or treatment options for CIPN, highlighting the need for the discovery of novel therapeutics and improving our understanding of disease mechanisms. In this study, we utilized human-induced pluripotent stem cell (hiPSC)-derived motor neurons as a platform to mimic axonal damage after treatment with vincristine, a chemotherapeutic used for the treatment of breast cancers, osteosarcomas, and leukemia. We screened a total of 1902 small molecules for neuroprotective properties in rescuing vincristine-induced axon growth deficits. From our primary screen, we identified 38 hit compounds that were subjected to secondary dose response screens. Six compounds showed favorable pharmacological profiles - AZD7762, A-674563, Blebbistatin, Glesatinib, KW-2449, and Pelitinib, all novel neuroprotectants against vincristine toxicity to neurons. In addition, four of these six compounds also showed efficacy against vincristine-induced growth arrest in human iPSC-derived sensory neurons. In this study, we utilized high-throughput screening of a large library of compounds in a therapeutically relevant assay. We identified several novel compounds that are efficacious in protecting different neuronal subtypes from the toxicity induced by a common chemotherapeutic agent, vincristine which could have therapeutic potential in the clinic.
Collapse
Affiliation(s)
| | | | | | - Shannon Zhen
- Boston Childrens Hospital: Boston Children's Hospital
| | - Bhagat Singh
- Boston Childrens Hospital: Boston Children's Hospital
| | | | | | - Anya Cheng
- Boston Childrens Hospital: Boston Children's Hospital
| | | | | | - Jennifer A Smith
- Harvard Medical School Center for Blood Research: Harvard Medical School
| | | |
Collapse
|
8
|
Allison RL, Welby E, Ehlers V, Burand A, Isaeva O, Nieves Torres D, Highland J, Brandow AM, Stucky CL, Ebert AD. Sickle cell disease iPSC-derived sensory neurons exhibit increased excitability and sensitization to patient plasma. Blood 2024; 143:2037-2052. [PMID: 38427938 PMCID: PMC11143522 DOI: 10.1182/blood.2023022591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/25/2024] [Accepted: 02/14/2024] [Indexed: 03/03/2024] Open
Abstract
ABSTRACT Individuals living with sickle cell disease (SCD) experience severe recurrent acute and chronic pain. Challenges to gaining mechanistic insight into pathogenic SCD pain processes include differential gene expression and function of sensory neurons between humans and mice with SCD, and extremely limited availability of neuronal tissues from patients with SCD. Here, we used induced pluripotent stem cells (iPSCs), derived from patients with SCD, differentiated into sensory neurons (SCD iSNs) to begin to overcome these challenges. We characterize key gene expression and function of SCD iSNs to establish a model to investigate intrinsic and extrinsic factors that may contribute to SCD pain. Despite similarities in receptor gene expression, SCD iSNs show pronounced excitability using patch clamp electrophysiology. Furthermore, we find that plasma taken from patients with SCD during acute pain associated with a vaso-occlusive event increases the calcium responses to the nociceptive stimulus capsaicin in SCD iSNs compared with those treated with paired plasma from patients with SCD at steady state baseline or healthy control plasma samples. We identified high levels of the polyamine spermine in baseline and acute pain states of plasma from patients with SCD, which sensitizes SCD iSNs to subthreshold concentrations of capsaicin. Together, these data identify potential intrinsic mechanisms within SCD iSNs that may extend beyond a blood-based pathology.
Collapse
Affiliation(s)
- Reilly L. Allison
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Emily Welby
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Vanessa Ehlers
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Anthony Burand
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Olena Isaeva
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Damaris Nieves Torres
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - Janelle Highland
- Department of Pediatrics, Section of Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI
| | - Amanda M. Brandow
- Department of Pediatrics, Section of Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI
| | - Cheryl L. Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
9
|
Kim H, Roh D, Oh SB. EGFR Tyrosine Kinase Inhibitor Lazertinib Activates a Subset of Mouse Sensory Neurons Via TRPA1. THE JOURNAL OF PAIN 2024; 25:104435. [PMID: 38008390 DOI: 10.1016/j.jpain.2023.11.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/21/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
Lazertinib (JNJ-73841937, YH25448) is a mutant-selective irreversible epidermal growth factor receptor tyrosine kinase inhibitor targeting both the T790M and activating mutation while sparing wild-type epidermal growth factor receptor. Paresthesia is one of the most common adverse events seen with lazertinib treatment, suggesting that lazertinib could affect the sensory nervous system. However, the mechanism of action for this paresthesia remains unclear. In this study, we investigated whether and how lazertinib affects peripheral sensory neurons. Through Fura-2-based calcium imaging and whole-cell patch clamp recording in primary-cultured dorsal root ganglion (DRG) neurons from adult mice, we found that application of lazertinib elicits spontaneous calcium responses in a subset of small-to-medium-sized neurons. Moreover, lazertinib induced spontaneous firings and hyperexcitability in a subset of transient receptor potential vanilloid 1-lineage DRG neurons and sensitized transient receptor potential ankyrin 1 (TRPA1) response, while sparing transient receptor potential vanilloid 1 response. Lazertinib-responsive neurons were also responsive to capsaicin, further supporting that lazertinib selectively activates nociceptive neurons. Lazertinib-induced calcium responses were pharmacologically blocked with HC-030031 (TRPA1 antagonist) and MDL-12330A (adenylyl cyclase inhibitor), suggesting that lazertinib activates sensory neurons through indirect activation of TRPA1. However, unlike vincristine which produces peripheral neuropathy by axonal degeneration, lazertinib did not cause neurite fragmentation in cultured DRG neurons. Finally, intraplantar injection of lazertinib induced TRPA1-dependent pain-like behaviors in vivo. Collectively, our data suggest a direct effect of lazertinib on nociceptive sensory neurons via TRPA1 selective mechanisms, which could be a putative mechanism of lazertinib-induced sensory abnormalities in clinical patients. PERSPECTIVE: This article presents a TRPA1-dependent, lazertinib-induced activation of mouse sensory neurons in vitro and lazertinib-induced pain-like behaviors in vivo. The same mechanisms may underlie the clinical condition, suggesting that TRPA1 could be a potential therapeutic target to manage lazertinib-induced paresthesia.
Collapse
Affiliation(s)
- Hayun Kim
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul 08826, Republic of Korea
| | - Dahee Roh
- Department of Neurobiology and Physiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Seog Bae Oh
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul 08826, Republic of Korea; Department of Neurobiology and Physiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| |
Collapse
|
10
|
Gomez-Deza J, Slavutsky AL, Nebiyou M, Le Pichon CE. Local production of reactive oxygen species drives vincristine-induced axon degeneration. Cell Death Dis 2023; 14:807. [PMID: 38065950 PMCID: PMC10709426 DOI: 10.1038/s41419-023-06227-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 08/27/2023] [Accepted: 10/18/2023] [Indexed: 12/18/2023]
Abstract
Neurological side effects arising from chemotherapy, such as severe pain and cognitive impairment, are a major concern for cancer patients. These major side effects can lead to reduction or termination of chemotherapy medication in patients, negatively impacting their prognoses. With cancer survival rates improving dramatically, addressing side effects of cancer treatment has become pressing. Here, we use iPSC-derived human neurons to investigate the molecular mechanisms that lead to neurotoxicity induced by vincristine, a common chemotherapeutic used to treat solid tumors. Our results uncover a novel mechanism by which vincristine causes a local increase in mitochondrial proteins that produce reactive oxygen species (ROS) in the axon. Vincristine triggers a cascade of axon pathology, causing mitochondrial dysfunction that leads to elevated axonal ROS levels and SARM1-dependent axon degeneration. Importantly, we show that the neurotoxic effect of increased axonal ROS can be mitigated by the small molecule mitochondrial division inhibitor 1 (mdivi-1) and antioxidants glutathione and mitoquinone, identifying a novel therapeutic avenue to treat the neurological effects of chemotherapy.
Collapse
Affiliation(s)
- Jorge Gomez-Deza
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Anastasia L Slavutsky
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Matthew Nebiyou
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Claire E Le Pichon
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
11
|
Mortensen C, Steffensen KD, Simonsen E, Herskind K, Madsen JS, Olsen DA, Iversen DB, Bergmann TK, Pottegård A, Stage TB. Neurofilament light chain as a biomarker of axonal damage in sensory neurons and paclitaxel-induced peripheral neuropathy in patients with ovarian cancer. Pain 2023; 164:1502-1511. [PMID: 36508173 DOI: 10.1097/j.pain.0000000000002840] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/08/2022] [Indexed: 12/14/2022]
Abstract
ABSTRACT Paclitaxel-induced peripheral neuropathy (PIPN) is a barrier to effective cancer treatment and impacts quality of life among patients with cancer. We used a translational approach to assess the utility of neurofilament light chain (NFL) as a biomarker of PIPN in a human cell model and in patients with ovarian cancer. We measured NFL in medium from human induced pluripotent stem cell-derived sensory neurons (iPSC-SNs) exposed to paclitaxel. Serum NFL (sNFL) levels were quantified in 190 patients with ovarian cancer receiving paclitaxel/carboplatin chemotherapy at baseline and after each of the following 2 or 6 cycles. Adverse outcomes related to PIPN were retrospectively obtained, and Cox regression model was performed with different sNFL cut-offs after first cycle. The apparent elimination half-life of sNFL was estimated in patients who discontinued paclitaxel. Paclitaxel neurotoxicity in iPSC-SNs was accompanied by NFL release in a concentration-dependent manner ( P < 0.001, analysis of variance). Serum NFL levels increased substantially in patients during paclitaxel/carboplatin chemotherapy with considerable interindividual variability. Patients with sNFL >150 pg/mL after first cycle had increased risk to discontinue paclitaxel early (unadjusted HR: 2.47 [95% CI 1.16-5.22], adjusted HR: 2.25 [95% CI: 0.88-5.79]). Similar trends were shown for risk of severe PIPN and paclitaxel dose reduction because of PIPN. The median elimination half-life of sNFL was 43 days (IQR 27-82 days). Neurofilament light chain constitutes an objective biomarker of neurotoxicity in iPSC-SNs and in ovarian cancer patients with high sNFL predicting PIPN-related adverse outcomes. If prospectively validated, NFL can be used to study PIPN and may guide clinical decision making and personalize treatment with paclitaxel.
Collapse
Affiliation(s)
- Christina Mortensen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Karina Dahl Steffensen
- Department of Oncology, Lillebaelt University Hospital of Southern Denmark, Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Emma Simonsen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Kamille Herskind
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Jonna Skov Madsen
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Biochemistry and Immunology, Lillebaelt University Hospital of Southern Denmark, Vejle, Denmark
| | - Dorte Aalund Olsen
- Department of Biochemistry and Immunology, Lillebaelt University Hospital of Southern Denmark, Vejle, Denmark
| | - Ditte Bork Iversen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Troels Korshøj Bergmann
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Pharmacology, Odense University Hospital, Odense, Denmark
| | - Anton Pottegård
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Tore Bjerregaard Stage
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
- Department of Clinical Pharmacology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
12
|
Suzuki I, Matsuda N, Han X, Noji S, Shibata M, Nagafuku N, Ishibashi Y. Large-Area Field Potential Imaging Having Single Neuron Resolution Using 236 880 Electrodes CMOS-MEA Technology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2207732. [PMID: 37088859 PMCID: PMC10369302 DOI: 10.1002/advs.202207732] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/21/2023] [Indexed: 05/03/2023]
Abstract
The electrophysiological technology having a high spatiotemporal resolution at the single-cell level and noninvasive measurements of large areas provide insights on underlying neuronal function. Here, a complementary metal-oxide semiconductor (CMOS)-microelectrode array (MEA) is used that uses 236 880 electrodes each with an electrode size of 11.22 × 11.22 µm and 236 880 covering a wide area of 5.5 × 5.9 mm in presenting a detailed and single-cell-level neural activity analysis platform for brain slices, human iPS cell-derived cortical networks, peripheral neurons, and human brain organoids. Propagation pattern characteristics between brain regions changes the synaptic propagation into compounds based on single-cell time-series patterns, classification based on single DRG neuron firing patterns and compound responses, axonal conduction characteristics and changes to anticancer drugs, and network activities and transition to compounds in brain organoids are extracted. This detailed analysis of neural activity at the single-cell level using the CMOS-MEA provides a new understanding of the basic mechanisms of brain circuits in vitro and ex vivo, on human neurological diseases for drug discovery, and compound toxicity assessment.
Collapse
Affiliation(s)
- Ikuro Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Naoki Matsuda
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Xiaobo Han
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Shuhei Noji
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Mikako Shibata
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Nami Nagafuku
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Yuto Ishibashi
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| |
Collapse
|
13
|
Chang YC, Lo YC, Chang HS, Lin HC, Chiu CC, Chen YF. An efficient cellular image-based platform for high-content screening of neuroprotective agents against chemotherapy-induced neuropathy. Neurotoxicology 2023; 96:118-128. [PMID: 37086979 DOI: 10.1016/j.neuro.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/12/2023] [Accepted: 04/19/2023] [Indexed: 04/24/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a major dose-limiting side effect, with no approved therapy for prevention or treatment. Here, we aimed to establish a high-content image platform based on the neurite outgrowth of dorsal root ganglia (DRG)-derived neuron cells for the discovery of neuroprotective agents against paclitaxel-induced CIPN. ND7/23 cells, an immortalized hybrid DRG cell line, were maturely differentiated by induction with nerve growth factor and upregulation of intracellular cAMP levels. High-content image analyses of the neurofilament-stained neurite network showed that paclitaxel disrupted the neurite outgrowth of well-differentiated ND7/23 DRG neuron cells, recapitulating characteristic effects of paclitaxel on primary cultured DRG neurons. This process coincided with the upregulated activity of store-operated Ca2+ entry, similar to those found in rodent models of paclitaxel-induced CIPN. The previously identified neuroprotective agents, minoxidil and 8-Br-cyclic adenosine monophosphate ribose (8-Br-cADPR), attenuated the reduction in total neurite outgrowth in paclitaxel-damaged ND7/23 cells. Additionally, the total neurite outgrowth of well-differentiated ND7/23 cells was concentration-dependently reduced by the neurotoxic chemotherapeutic agents, oxaliplatin and bortezomib, but not the less neurotoxic 5-fluorouracil. We demonstrated that high-content analyses of neurite morphology in well-differentiated DRG neuron-derived cells provide an effective, reproducible, and high-throughput strategy for developing therapeutics against CIPN.
Collapse
Affiliation(s)
- Yang-Chen Chang
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Ching Lo
- Department of Pharmacology, School of Medicine, Kaohsiung Medical University, Taiwan
| | - Hsun-Shuo Chang
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Hui-Ching Lin
- Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, School of Life Science, Kaohsiung Medical University, Taiwan
| | - Yih-Fung Chen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
14
|
Allison RL, Burand A, Torres DN, Brandow AM, Stucky CL, Ebert AD. Sickle cell disease patient plasma sensitizes iPSC-derived sensory neurons from sickle cell disease patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.10.523446. [PMID: 36711992 PMCID: PMC9882050 DOI: 10.1101/2023.01.10.523446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Individuals living with sickle cell disease (SCD) experience severe recurrent acute and chronic pain. In order to develop novel therapies, it is necessary to better understand the neurobiological mechanisms underlying SCD pain. There are many barriers to gaining mechanistic insight into pathogenic SCD pain processes, such as differential gene expression and function of sensory neurons between humans and mice with SCD, as well as the limited availability of patient samples. These can be overcome by utilizing SCD patient-derived induced pluripotent stem cells (iPSCs) differentiated into sensory neurons (SCD iSNs). Here, we characterize the key gene expression and function of SCD iSNs to establish a model for higher-throughput investigation of intrinsic and extrinsic factors that may contribute to increased SCD patient pain. Importantly, identified roles for C-C Motif Chemokine Ligand 2 (CCL2) and endothelin 1 (ET1) in SCD pain can be recapitulated in SCD iSNs. Further, we find that plasma taken from SCD patients during acute pain increases SCD iSN calcium response to the nociceptive stimulus capsaicin compared to those treated with paired SCD patient plasma at baseline or healthy control plasma samples. Together, these data provide the framework necessary to utilize iSNs as a powerful tool to investigate the neurobiology of SCD and identify potential intrinsic mechanisms of SCD pain which may extend beyond a blood-based pathology.
Collapse
Affiliation(s)
- Reilly L. Allison
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Anthony Burand
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Damaris Nieves Torres
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - Amanda M. Brandow
- Department of Pediatrics, Section of Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI
| | - Cheryl L. Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
15
|
Gao Y. Using Human iPSC-Derived Peripheral Nervous System Disease Models for Drug Discovery. Handb Exp Pharmacol 2023; 281:191-205. [PMID: 37815594 DOI: 10.1007/164_2023_690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
Induced pluripotent stem cells (IPSCs), with their remarkable ability to differentiate into various cell types, including peripheral nervous system cells such as neurons and glial cells, offer an excellent platform for in vitro disease modeling. These iPSC-derived disease models have proven valuable in drug discovery, as they provide more precise simulations of a patient's disease state and allow for the assessment of potential therapeutic effectiveness and safety.
Collapse
Affiliation(s)
- Yuan Gao
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
16
|
Richter MN, Fattahi F. Stem Cell-Based Models for Studying the Effects of Cancer and Cancer Therapies on the Peripheral Nervous System. Adv Biol (Weinh) 2022; 6:e2200009. [PMID: 35666079 DOI: 10.1002/adbi.202200009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/06/2022] [Indexed: 01/28/2023]
Abstract
In recent years, the complexity of cancer and cancer therapies and their interactions with the peripheral nervous system have come into focus, but limitations in experimental models have remained a significant challenge in the field. As evidence, there are currently no therapies approved that target cancer-peripheral nervous system or cancer therapy-peripheral nervous system interactions as an anti-neoplastic or anti-neurotoxic agent, respectively. Human pluripotent stem cells offer an appealing model system that, unlike rodent models, is compatible with high throughput, high content applications; techniques that reflect modern drug discovery methodologies. Thus, utilizing the key advantages of stem cell-based models in tandem with the strengths of traditional animal models offers a complementary and interdisciplinary strategy to advance cancer and cancer therapy-peripheral nervous system research and drug discovery. In this review, the current status of the cancer-peripheral nervous system and cancer therapy-peripheral nervous system research is discussed, examples where stem cell-based models have been implemented are described, and avenues where stem cell-based models may further advance the field are proposed.
Collapse
Affiliation(s)
- Mikayla N Richter
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, 94158, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA
| | - Faranak Fattahi
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, 94158, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA
- Program in Craniofacial Biology, University of California, San Francisco, CA, 94110, USA
| |
Collapse
|
17
|
Mortensen C, Andersen NE, Stage TB. Bridging the Translational Gap in Chemotherapy-Induced Peripheral Neuropathy with iPSC-Based Modeling. Cancers (Basel) 2022; 14:cancers14163939. [PMID: 36010931 PMCID: PMC9406154 DOI: 10.3390/cancers14163939] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Chemotherapy-induced peripheral neuropathy (CIPN) remains a clinical challenge with a considerable impact on the effective treatment of cancers and quality of life during and after concluding chemotherapy. Given the limited understanding of CIPN, there are no options for the treatment and prevention of CIPN. Decades of research with the unsuccessful translation of preclinical findings to clinical studies argue for the requirement of human model systems. This review focuses on the translational potential of human induced pluripotent stem cells (iPSCs) in CIPN research. We provide an overview of the current studies and discuss important aspects to improve the translation of in vitro findings. We identified distinct effects on the neurite network and cell viability upon exposure to different classes of chemotherapy. Our study revealed considerable variability between donors and between neurons of the central and peripheral nervous system. Translational success may be improved by including multiple iPSC donors with known clinical data and selecting clinically relevant concentrations. Abstract Chemotherapy-induced peripheral neuropathy (CIPN) is a common and potentially serious adverse effect of a wide range of chemotherapeutics. The lack of understanding of the molecular mechanisms underlying CIPN limits the efficacy of chemotherapy and development of therapeutics for treatment and prevention of CIPN. Human induced pluripotent stem cells (iPSCs) have become an important tool to generate the cell types associated with CIPN symptoms in cancer patients. We reviewed the literature for iPSC-derived models that assessed neurotoxicity among chemotherapeutics associated with CIPN. Furthermore, we discuss the gaps in our current knowledge and provide guidance for selecting clinically relevant concentrations of chemotherapy for in vitro studies. Studies in iPSC-derived neurons revealed differential sensitivity towards mechanistically diverse chemotherapeutics associated with CIPN. Additionally, the sensitivity to chemotherapy was determined by donor background and whether the neurons had a central or peripheral nervous system identity. We propose to utilize clinically relevant concentrations that reflect the free, unbound fraction of chemotherapeutics in plasma in future studies. In conclusion, iPSC-derived sensory neurons are a valuable model to assess CIPN; however, studies in Schwann cells and motor neurons are warranted. The inclusion of multiple iPSC donors and concentrations of chemotherapy known to be achievable in patients can potentially improve translational success.
Collapse
Affiliation(s)
- Christina Mortensen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, DK-5000 Odense C, Denmark
| | - Nanna Elman Andersen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, DK-5000 Odense C, Denmark
| | - Tore Bjerregaard Stage
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, DK-5000 Odense C, Denmark
- Department of Clinical Pharmacology, Odense University Hospital, DK-5000 Odense C, Denmark
- Correspondence:
| |
Collapse
|
18
|
Liu B, Kong Y, Shi W, Kuss M, Liao K, Hu G, Xiao P, Sankarasubramanian J, Guda C, Wang X, Lei Y, Duan B. Exosomes derived from differentiated human ADMSC with the Schwann cell phenotype modulate peripheral nerve-related cellular functions. Bioact Mater 2022; 14:61-75. [PMID: 35310346 PMCID: PMC8892082 DOI: 10.1016/j.bioactmat.2021.11.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/09/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Peripheral nerve regeneration remains a significant clinical challenge due to the unsatisfactory functional recovery and public health burden. Exosomes, especially those derived from mesenchymal stem cells (MSCs), are promising as potential cell-free therapeutics and gene therapy vehicles for promoting neural regeneration. In this study, we reported the differentiation of human adipose derived MSCs (hADMSCs) towards the Schwann cell (SC) phenotype (hADMSC-SCs) and then isolated exosomes from hADMSCs with and without differentiation (i.e., dExo vs uExo). We assessed and compared the effects of uExo and dExo on antioxidative, angiogenic, anti-inflammatory, and axon growth promoting properties by using various peripheral nerve-related cells. Our results demonstrated that hADMSC-SCs secreted more neurotrophic factors and other growth factors, compared to hADMSCs without differentiation. The dExo isolated from hADMSC-SCs protected rat SCs from oxidative stress and enhanced HUVEC migration and angiogenesis. Compared to uExo, dExo also had improved performances in downregulating pro-inflammatory gene expressions and cytokine secretions and promoting axonal growth of sensory neurons differentiated from human induced pluripotent stem cells. Furthermore, microRNA (miRNA) sequencing analysis revealed that exosomes and their parent cells shared some similarities in their miRNA profiles and exosomes displayed a distinct miRNA signature. Many more miRNAs were identified in dExo than in uExo. Several upregulated miRNAs, like miRNA-132-3p and miRNA-199b-5p, were highly related to neuroprotection, anti-inflammation, and angiogenesis. The dExo can effectively modulate various peripheral nerve-related cellular functions and is promising for cell-free biological therapeutics to enhance neural regeneration. Exosomes were isolated from hADMSCs with and without differentiation towards SC phenotype (i.e., dExo vs uExo). hADMSC-SCs secreted more growth factors compared to hADMSCs without differentiation. The dExo protected rat SCs from oxidative stress and enhanced endothelial cell migration and angiogenesis. dExo promoted axonal growth of sensory neurons differentiated from hiPSCs. miRNA sequencing analysis unveiled and compared the exosomal and cellular miRNA profiles.
Collapse
|
19
|
Kumar Kalvala A, Bagde A, Arthur P, Kumar Surapaneni S, Ramesh N, Nathani A, Singh M. Role of Cannabidiol and Tetrahydrocannabivarin on Paclitaxel-induced neuropathic pain in rodents. Int Immunopharmacol 2022; 107:108693. [PMID: 35303507 PMCID: PMC10791145 DOI: 10.1016/j.intimp.2022.108693] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/26/2022] [Accepted: 03/07/2022] [Indexed: 01/13/2023]
Abstract
The purpose of this study was to evaluate if phytocannabinoids, synthetic cannabidiol (CBD), and tetrahydrocannabivarin (THCV), and their combination, could protect mice from Paclitaxel-induced peripheral neuropathy (PIPN). Six groups of C57BL/6J mice (n = 6) were used in this study. The mice were given paclitaxel (PTX) (8 mg/kg/day, i.p.) on days 1, 3, 5, and 7 to induce neuropathy. Mice were evaluated for behavioral parameters, and dorsal root ganglions (DRG) were collected from the animals and subjected to RNA sequencing and westernblot analysis at the end of the study. On cultured DRGs derived from adult male rats, immunocytochemistry and mitochondrial functional assays were also performed. When compared to individual treatments, the combination of CBD and THCV improved thermal and mechanical neurobehavioral symptoms in mice by twofold. Targets for CBD and THCV therapy were identified by KEGG (RNA sequencing). PTX reduced the expression of p-AMPK, SIRT1, NRF2, HO1, SOD2, and catalase while increasing the expression of PI3K, p-AKT, p-P38 MAP kinase, BAX, TGF-β, NLRP3 inflammasome, and caspase 3 in DRG homogenates of mice. Combination therapy outperformed monotherapy in reversing these protein expressions. The addition of CBD and THCV to DRG primary cultures reduced mitochondrial superoxides while increasing mitochondrial membrane potentials. WAY100135 and rimonabant altered the neuroprotective effects of CBD and THCV respectively by blocking 5-HT1A and CB1 receptors in mice and DRG primary cultures. The entourage effect of CBD and THCV against PIPN appears to protect neurons in mice via 5HT1A and CB1 receptors respectively.
Collapse
Affiliation(s)
- Anil Kumar Kalvala
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Arvind Bagde
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Peggy Arthur
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Sunil Kumar Surapaneni
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Nimma Ramesh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA.
| |
Collapse
|
20
|
Huehnchen P, Schinke C, Bangemann N, Dordevic AD, Kern J, Maierhof SK, Hew L, Nolte L, Körtvelyessy P, Göpfert JC, Ruprecht K, Somps CJ, Blohmer JU, Sehouli J, Endres M, Boehmerle W. Neurofilament proteins as potential biomarker in chemotherapy-induced polyneuropathy. JCI Insight 2022; 7:154395. [PMID: 35133982 PMCID: PMC8986065 DOI: 10.1172/jci.insight.154395] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 02/02/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Paclitaxel chemotherapy frequently induces dose-limiting sensory axonal polyneuropathy. As sensory symptoms are challenging to assess objectively in clinical routine, an easily accessible biomarker for chemotherapy-induced polyneuropathy (CIPN) holds the potential to improve early diagnosis. Here, we describe neurofilament light chain (NFL), a marker for neuroaxonal damage, as translational surrogate marker for CIPN. METHODS NFL concentrations were measured in an in vitro model of CIPN, exposing induced pluripotent stem cell-derived sensory neurons (iPSC-DSN) to paclitaxel. Breast and ovarian cancer patients undergoing paclitaxel chemotherapy, breast cancer control patients without chemotherapy and healthy controls were recruited in a cohort study and examined before chemotherapy (V1) and after 28 weeks (V2, after chemotherapy). CIPN was assessed by the validated Total Neuropathy Score reduced, which combines patient-reported symptoms with data from clinical examinations. Serum NFL (NFLs) concentrations were measured at both visits with single molecule array technology (SIMOA). RESULTS NFL is released from iPSC-DSN upon paclitaxel incubation in a dose- and time-dependent manner and inversely correlates with iPSC-DSN viability. NFLs strongly increased in paclitaxel-treated patients with CIPN, but not in chemotherapy patients without CIPN or controls, resulting in an 86 % sensitivity and 87 % specificity. A NFLs increase of +36 pg/ml from baseline was associated with a predicted CIPN probability of >0.5. CONCLUSION NFLs correlates with CIPN development and severity, which may guide neurotoxic chemotherapy in the future. TRIAL REGISTRATION NCT02753036FUNDING. DFG (EXC 257 NeuroCure), BMBF (01 EO 0801), AnimalFreeResearch Organization, EU Horizon 2020 Innovative Medicines Initiative 2 Joint Undertaking (TransBioLine, 821283).
Collapse
Affiliation(s)
- Petra Huehnchen
- Department of Experimental Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Schinke
- Department of Experimental Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Nikola Bangemann
- Gynecology and Systemic Gynecology, Carl-Thiem-Klinikum Cottbus, Cottbus, Germany
| | - Adam D Dordevic
- Department of Experimental Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Kern
- Department of Experimental Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Smilla K Maierhof
- Department of Experimental Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Lois Hew
- Department of Experimental Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Luca Nolte
- Department of Experimental Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Körtvelyessy
- Department of Experimental Neurology, Charite Universitätsmedizin Berlin, Berlin, Germany
| | - Jens C Göpfert
- Naturwissenschaftliches und Medizinisches Institut, Universität Tübingen, Reutlingen, Germany
| | - Klemens Ruprecht
- Department of Experimental Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Christopher J Somps
- Drug Safety Research and Development, Pfizer, Groton, United States of America
| | - Jens-Uwe Blohmer
- Department of Gynecology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jalid Sehouli
- Department of Gynecology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Endres
- Department of Experimental Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang Boehmerle
- Department of Experimental Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
21
|
Alsaloum M, Waxman SG. iPSCs and DRGs: stepping stones to new pain therapies. Trends Mol Med 2022; 28:110-122. [PMID: 34933815 PMCID: PMC8810720 DOI: 10.1016/j.molmed.2021.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 02/03/2023]
Abstract
There is a pressing need for more effective nonaddictive treatment options for pain. Pain signals are transmitted from the periphery into the spinal cord via dorsal root ganglion (DRG) neurons, whose excitability is driven by voltage-gated sodium (NaV) channels. Three NaV channels (NaV1.7, NaV1.8, and NaV1.9), preferentially expressed in DRG neurons, play important roles in pain signaling in humans. Blockade of these channels may provide a novel approach to the treatment of pain, but clinical translation of preclinical results has been challenging, in part due to differences between rodent and human DRG neurons. Human DRG neurons and iPSC-derived sensory neurons (iPSC-SNs) provide new preclinical platforms that may facilitate the development of novel pain therapeutics.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA; Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA; Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA; Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
22
|
Sadler KE, Mogil JS, Stucky CL. Innovations and advances in modelling and measuring pain in animals. Nat Rev Neurosci 2022; 23:70-85. [PMID: 34837072 PMCID: PMC9098196 DOI: 10.1038/s41583-021-00536-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2021] [Indexed: 12/12/2022]
Abstract
Best practices in preclinical algesiometry (pain behaviour testing) have shifted over the past decade as a result of technological advancements, the continued dearth of translational progress and the emphasis that funding institutions and journals have placed on rigour and reproducibility. Here we describe the changing trends in research methods by analysing the methods reported in preclinical pain publications from the past 40 years, with a focus on the last 5 years. We also discuss how the status quo may be hampering translational success. This discussion is centred on four fundamental decisions that apply to every pain behaviour experiment: choice of subject (model organism), choice of assay (pain-inducing injury), laboratory environment and choice of outcome measures. Finally, we discuss how human tissues, which are increasingly accessible, can be used to validate the translatability of targets and mechanisms identified in animal pain models.
Collapse
Affiliation(s)
- Katelyn E Sadler
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jeffrey S Mogil
- Department of Psychology, McGill University, Montreal, QC, Canada
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Cheryl L Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
23
|
Chua KC, El-Haj N, Priotti J, Kroetz DL. Mechanistic insights into the pathogenesis of microtubule-targeting agent-induced peripheral neuropathy from pharmacogenetic and functional studies. Basic Clin Pharmacol Toxicol 2022; 130 Suppl 1:60-74. [PMID: 34481421 PMCID: PMC8716520 DOI: 10.1111/bcpt.13654] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/04/2021] [Accepted: 09/01/2021] [Indexed: 01/03/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a common dose-limiting toxicity that affects 30%-40% of patients undergoing cancer treatment. Although multiple mechanisms of chemotherapy-induced neurotoxicity have been described in preclinical models, these have not been translated into widely effective strategies for the prevention or treatment of CIPN. Predictive biomarkers to inform therapeutic approaches are also lacking. Recent studies have examined genetic risk factors associated with CIPN susceptibility. This review provides an overview of the clinical and pathologic features of CIPN and summarizes efforts to identify target pathways through genetic and functional studies. Structurally and mechanistically diverse chemotherapeutics are associated with CIPN; however, the current review is focused on microtubule-targeting agents since these are the focus of most pharmacogenetic association and functional studies of CIPN. Genome-wide pharmacogenetic association studies are useful tools to identify not only causative genes and genetic variants but also genetic networks implicated in drug response or toxicity and have been increasingly applied to investigations of CIPN. Induced pluripotent stem cell-derived models of human sensory neurons are especially useful to understand the mechanistic significance of genomic findings. Combined genetic and functional genomic efforts to understand CIPN hold great promise for developing therapeutic approaches for its prevention and treatment.
Collapse
Affiliation(s)
- Katherina C. Chua
- Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California San Francisco, San Francisco, CA 94143-2911,Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143-2911
| | - Nura El-Haj
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143-2911
| | - Josefina Priotti
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143-2911
| | - Deanna L. Kroetz
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143-2911,Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143-2911
| |
Collapse
|
24
|
Cunningham GM, Shen F, Wu X, Cantor EL, Gardner L, Philips S, Jiang G, Bales CL, Tan Z, Liu Y, Wan J, Fehrenbacher JC, Schneider BP. The impact of SBF2 on taxane-induced peripheral neuropathy. PLoS Genet 2022; 18:e1009968. [PMID: 34986146 PMCID: PMC8765656 DOI: 10.1371/journal.pgen.1009968] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 01/18/2022] [Accepted: 11/26/2021] [Indexed: 12/13/2022] Open
Abstract
Taxane-induced peripheral neuropathy (TIPN) is a devastating survivorship issue for many cancer patients. In addition to its impact on quality of life, this toxicity may lead to dose reductions or treatment discontinuation, adversely impacting survival outcomes and leading to health disparities in African Americans (AA). Our lab has previously identified deleterious mutations in SET-Binding Factor 2 (SBF2) that significantly associated with severe TIPN in AA patients. Here, we demonstrate the impact of SBF2 on taxane-induced neuronal damage using an ex vivo model of SBF2 knockdown of induced pluripotent stem cell-derived sensory neurons. Knockdown of SBF2 exacerbated paclitaxel changes to cell viability and neurite outgrowth while attenuating paclitaxel-induced sodium current inhibition. Our studies identified paclitaxel-induced expression changes specific to mature sensory neurons and revealed candidate genes involved in the exacerbation of paclitaxel-induced phenotypes accompanying SBF2 knockdown. Overall, these findings provide ex vivo support for the impact of SBF2 on the development of TIPN and shed light on the potential pathways involved.
Collapse
Affiliation(s)
- Geneva M. Cunningham
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Fei Shen
- Department of Hematology and Oncology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Xi Wu
- Department of Hematology and Oncology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Erica L. Cantor
- Department of Hematology and Oncology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Laura Gardner
- Department of Hematology and Oncology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Santosh Philips
- Department of Clinical Pharmacology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Guanglong Jiang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Casey L. Bales
- Department of Clinical Pharmacology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Zhiyong Tan
- Department of Pharmacology and Toxicology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Jill C. Fehrenbacher
- Department of Pharmacology and Toxicology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| | - Bryan P. Schneider
- Department of Medical and Molecular Genetics, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
- Department of Hematology and Oncology, Indiana University School of Medicine; Indianapolis, Indiana, United States of America
| |
Collapse
|
25
|
Considerations for a Reliable In Vitro Model of Chemotherapy-Induced Peripheral Neuropathy. TOXICS 2021; 9:toxics9110300. [PMID: 34822690 PMCID: PMC8620674 DOI: 10.3390/toxics9110300] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 12/13/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is widely recognized as a potentially severe toxicity that often leads to dose reduction or discontinuation of cancer treatment. Symptoms may persist despite discontinuation of chemotherapy and quality of life can be severely compromised. The clinical symptoms of CIPN, and the cellular and molecular targets involved in CIPN, are just as diverse as the wide variety of anticancer agents that cause peripheral neurotoxicity. There is an urgent need for extensive molecular and functional investigations aimed at understanding the mechanisms of CIPN. Furthermore, a reliable human cell culture system that recapitulates the diversity of neuronal modalities found in vivo and the pathophysiological changes that underlie CIPN would serve to advance the understanding of the pathogenesis of CIPN. The demonstration of experimental reproducibility in a human peripheral neuronal cell system will increase confidence that such an in vitro model is clinically useful, ultimately resulting in deeper exploration for the prevention and treatment of CIPN. Herein, we review current in vitro models with a focus on key characteristics and attributes desirable for an ideal human cell culture model relevant for CIPN investigations.
Collapse
|
26
|
Chrysostomidou L, Cooper AH, Weir GA. Cellular models of pain: New technologies and their potential to progress preclinical research. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2021; 10:100063. [PMID: 34977426 PMCID: PMC8683679 DOI: 10.1016/j.ynpai.2021.100063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 01/16/2023]
Abstract
Human sensory neurons can reduce the translational gap in analgesic development. Access to dorsal root ganglion (hDRG) neurons is increasing. Diverse sensory neuron subtypes can now be generated via stem cell technology. Advances of these technologies will improve our understanding of human nociception.
In vitro models fill a vital niche in preclinical pain research, allowing detailed study of molecular pathways, and in the case of humanised systems, providing a translational bridge between in vivo animal models and human patients. Significant advances in cellular technology available to basic pain researchers have occurred in the last decade, including developing protocols to differentiate sensory neuron-like cells from stem cells and greater access to human dorsal root ganglion tissue. In this review, we discuss the use of both models in preclinical pain research: What can a human sensory neuron in a dish tell us that rodent in vivo models cannot? How similar are these models to their endogenous counterparts, and how should we judge them? What limitations do we need to consider? How can we leverage cell models to improve translational success? In vitro human sensory neuron models equip pain researchers with a valuable tool to investigate human nociception. With continual development, consideration for their advantages and limitations, and effective integration with other experimental strategies, they could become a driving force for the pain field's advancement.
Collapse
Affiliation(s)
- Lina Chrysostomidou
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Andrew H Cooper
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Greg A Weir
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
27
|
Schinke C, Fernandez Vallone V, Ivanov A, Peng Y, Körtvelyessy P, Nolte L, Huehnchen P, Beule D, Stachelscheid H, Boehmerle W, Endres M. Modeling chemotherapy induced neurotoxicity with human induced pluripotent stem cell (iPSC) -derived sensory neurons. Neurobiol Dis 2021; 155:105391. [PMID: 33984509 DOI: 10.1016/j.nbd.2021.105391] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/20/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a frequent, potentially irreversible adverse effect of cytotoxic chemotherapy often leading to a reduction or discontinuation of treatment which negatively impacts patients' prognosis. To date, however, neither predictive biomarkers nor preventive treatments for CIPN are available, which is partially due to a lack of suitable experimental models. We therefore aimed to evaluate whether sensory neurons derived from induced pluripotent stem cells (iPSC-DSN) can serve as human disease model system for CIPN. Treatment of iPSC-DSN for 24 h with the neurotoxic drugs paclitaxel, bortezomib, vincristine and cisplatin led to axonal blebbing and a dose dependent decline of cell viability in clinically relevant IC50 ranges, which was not observed for the non-neurotoxic compounds doxorubicin and 5-fluorouracil. Paclitaxel treatment effects were less pronounced after 24 h but prominent when treatment was applied for 72 h. Global transcriptome analyses performed at 24 h, i.e. before paclitaxel-induced cell death occurred, revealed the differential expression of genes of neuronal injury, cellular stress response, and sterol pathways. We further evaluated if known neuroprotective strategies can be reproduced in iPSC-DSN and observed protective effects of lithium replicating findings from rodent dorsal root ganglia cells. Comparing sensory neurons derived from two different healthy donors, we found preliminary evidence that these cell lines react differentially to neurotoxic drugs as expected from the variable presentation of CIPN in patients. In conclusion, iPSC-DSN are a promising platform to study the pathogenesis of CIPN and to evaluate neuroprotective treatment strategies. In the future, the application of patient-specific iPSC-DSN could open new avenues for personalized medicine with individual risk prediction, choice of chemotherapeutic compounds and preventive treatments.
Collapse
Affiliation(s)
- Christian Schinke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178 Berlin, Germany
| | - Valeria Fernandez Vallone
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Stem Cell Core Facility, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Andranik Ivanov
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioinformatics, Charitéplatz 1, 10117 Berlin, Germany
| | - Yangfan Peng
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institut für Neurophysiologie, Charitéplatz 1, 10117 Berlin, Germany
| | - Péter Körtvelyessy
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, Charitéplatz 1, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases, 39120 Magdeburg, Germany
| | - Luca Nolte
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany
| | - Petra Huehnchen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany
| | - Dieter Beule
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioinformatics, Charitéplatz 1, 10117 Berlin, Germany; Max-Delbrueck Center for Molecular Medicine, 13125 Berlin, Germany
| | - Harald Stachelscheid
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Stem Cell Core Facility, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Wolfgang Boehmerle
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany.
| | - Matthias Endres
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), partner site Berlin, Germany; German Center for Cardiovascular Research (DZHK), partner site Berlin, Germany
| |
Collapse
|
28
|
Li Y, Chen X, Zhu Q, Chen R, Xu L, Li S, Shi X, Xu H, Xu Y, Zhang W, Huang X, Zha X, Wang J. Retrospective comparisons of nanoparticle albumin-bound paclitaxel and docetaxel neoadjuvant regimens for breast cancer. Nanomedicine (Lond) 2021; 16:391-400. [PMID: 33502252 DOI: 10.2217/nnm-2020-0458] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Aim: To compare the efficacy and safety of 2-weekly nanoparticle albumin-bound paclitaxel (nP) and 3-weekly docetaxel regimens as neoadjuvant systemic therapy (NST) for breast cancer. Materials & methods: Patients (n = 201) received NST comprising either dose-dense epirubicin and cyclophosphamide followed by 2-weekly nP (n = 104) or 3-weekly courses of epirubicin and cyclophosphamide followed by docetaxel (n = 97). Results: Higher pathological complete response rates were achieved by the nP group. Subgroup analysis showed that the nP-based regimen achieved higher pathological complete response rates in patients with triple-negative tumor cells and high Ki67 levels. However, grades 3-4 peripheral sensory neuropathies were more frequent in the nP group. Conclusion: The 2-weekly nP-based regimen might be a better choice of NST for patients with breast cancer.
Collapse
Affiliation(s)
- Yan Li
- Department of Breast Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, PR China
| | - Xiang Chen
- Department of Thyroid and Mammary Gland Surgery, Yixing People's Hospital, Wuxi 214200, PR China
| | - Qiannan Zhu
- Department of Breast Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, PR China
| | - Rui Chen
- Department of Breast Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, PR China
| | - Lu Xu
- Department of Clinical Nutrition, First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, PR China
| | - Shuo Li
- Department of Breast Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, PR China
| | - Xiaoqing Shi
- Department of Breast Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, PR China
| | - Haiping Xu
- Department of Breast Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, PR China
| | - Yinggang Xu
- Department of Breast Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, PR China
| | - Weiwei Zhang
- Department of Breast Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, PR China
| | - Xiaofeng Huang
- Department of Breast Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, PR China
| | - Xiaoming Zha
- Department of Breast Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, PR China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210000, PR China
| | - Jue Wang
- Department of Breast Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, PR China.,Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210000, PR China
| |
Collapse
|