1
|
Milheiro C, Moura ML, Amendola M, Barbosa MA, Caldeira J. Harnessing CRISPR potential for intervertebral disc regeneration strategies. Front Bioeng Biotechnol 2025; 13:1562412. [PMID: 40406584 PMCID: PMC12095242 DOI: 10.3389/fbioe.2025.1562412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/15/2025] [Indexed: 05/26/2025] Open
Abstract
Genome editing technologies, particularly CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats), have broadened the possibilities of genetic research and molecular biology by enabling precise modifications of the genome, offering novel therapeutic potential for various disorders. Herein, we present an overview of traditional genome editing techniques and delve deeper into the CRISPR toolbox, with particular attention given to epigenetic and transcriptional regulation. In the context of the intervertebral disc (IVD), CRISPR offers an unprecedented approach to address the mechanisms underlying tissue degeneration, advancing the development of revolutionary therapies for Low Back Pain (LBP). As so, we showcase how to leverage CRISPR systems for IVD. This cutting-edge technology has been successfully used to improve our understanding of IVD biology through functional studies and disease modeling. Most relevant research prioritizes new targets associated with the extracellular matrix (ECM), pain sensing or inflammatory pathways. Promising CRISPR applications encompass IVD regeneration by recapitulation of a regenerative environment or by targeting important degenerative catalysts. In the future, priority should be given to fetal gene reactivation, multiple healthy gene expression enhancement and disease-associated polymorphisms' correction. Despite several challenges such as effective delivery, off-target effects, as well as ethical and safety concerns, exciting clinical trials are anticipated in the years to come, providing more effective and long-lasting solutions for IVD degeneration.
Collapse
Affiliation(s)
- Catarina Milheiro
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Maria L. Moura
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| | - Mario Amendola
- Généthon, Évry, France
- Integrare Research Unit UMR_S951, Université Paris-Saclay, Université Evry, Inserm, Généthon, Évry, France
| | - Mário A. Barbosa
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Joana Caldeira
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| |
Collapse
|
2
|
Beall DP, Davis TT, Amirdelfan K, Naidu RK, DePalma MJ, Costandi S, Yoon ES, Fleming JW, Block JE, Mekhail N. Supplemental nucleus pulposus allograft in patients with lumbar discogenic pain: results of a prospective feasibility study. BMC Musculoskelet Disord 2025; 26:437. [PMID: 40312677 PMCID: PMC12044970 DOI: 10.1186/s12891-025-08701-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Degeneration of the intervertebral disc is a significant source of chronic axial low back pain. Direct supplementation of degenerated nucleus pulposis (NP) tissue with intradiscally delivered allogeneic NP represents an opportunity to bridge the treatment gap between failed conservative care and spine surgery for patients with lumbar discogenic pain. METHODS Prospective, single-arm clinical study conducted at 6 sites in the US. The primary objective was to determine the magnitude of improvement in back pain severity and back function in patients with chronic lumbar discogenic pain at 12 months after a single intradiscal supplementation procedure using a commercially available NP allograft at up to two vertebral levels identified on magnetic resonance imaging. Back pain severity was evaluated using an 11-point numeric rating scale (NRS) and back function using the Oswestry Disability Index (ODI). Minimal clinically important difference (MCID) and substantial clinical benefit (SCB) were set at ≥ 30% and ≥ 50% over baseline, respectively. The patient acceptable symptom state (PASS) threshold for pain severity was ≤ 3. RESULTS Twenty-eight participants with a mean age of 44 ± 13 yrs. were enrolled and 22 provided 12-month outcomes. The average overall improvement in back pain severity was 43% through 12 months (p < 0.001). Approximately 64% (14 of 22) achieved the MCID in back pain at 12 months, with 55% (12 of 22) realizing SCB. Almost 60% (13 of 22) reported a 12-month back pain severity score of ≤ 3. The corresponding average decrease in ODI values was 50% (p < 0.001) with approximately 59% (13 of 22) of study participants achieving the MCID. At baseline approximately 82% (23 of 28) of participants reported severe or crippled back impairment compared to 18% (4 of 22) at 12 months (p < 0.001). CONCLUSION The results of this study provide additional evidence that supplementation of the degenerated intervertebral disc with intradiscally delivered allogeneic NP is associated with clinically significant pain palliation and functional improvement. TRIAL REGISTRATION This trial was prospectively registered at ClinicalTrials.gov on December 30, 2021 (NCT05201287).
Collapse
Affiliation(s)
- Douglas P Beall
- Comprehensive Specialty Care, 1023 Waterwood Pkwy, Edmond, OK, 73034, USA
| | - Timothy T Davis
- Source Healthcare, 2801 Wilshire Blvd, Ste. A, Santa Monica, CA, 90403, USA
| | - Kasra Amirdelfan
- Boomerang Healthcare, Inc, 450 N. Wiget Lane, Walnut Creek, CA, 94598, USA
| | - Ramana K Naidu
- MarinHealth Spine Institute, 2 Bon Air Road, Ste. 120, Larkspur, CA, 94939, USA
| | - Michael J DePalma
- Virginia iSpine Physicians, 12874 Patterson Avenue, Ste. A, Richmond, VA, 23238, USA
| | - Shrif Costandi
- Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Edward S Yoon
- Hospital for Special Surgery, 535 East 70 Street, New York, NY, 10021, USA
| | | | - Jon E Block
- , 2210 Jackson Street, Ste. 401, San Francisco, CA, 94115, USA.
| | - Nagy Mekhail
- Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| |
Collapse
|
3
|
Trone MAR, Stover JD, Almarza A, Bowles RD. pH: A major player in degenerative intervertebral disks. JOR Spine 2024; 7:e70025. [PMID: 39703199 PMCID: PMC11655178 DOI: 10.1002/jsp2.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/04/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024] Open
Abstract
Chronic lower back pain is the leading cause of disability worldwide, generating a socioeconomic cost of over $100 billion annually in the United States. Among the prominent causes of low back pain (LBP) is degeneration of the intervertebral disk (IVD), a condition known as degenerative disk disease (DDD). Despite the prevalence of DDD and multiple studies demonstrating its relationship with LBP, the mechanisms by which it contributes to pain remain unknown. Previous studies have identified potential causes for this pain, such as extracellular matrix (ECM) breakdown, changes in biomechanics, and pro-inflammatory signals. Possible pain treatments targeting these factors have been developed but with limited effects. However, low pH in DDD is a potential pain generator whose role has largely been unexplored and underappreciated. This review highlights hyperacidity's effects on the IVD, such as catabolism of disk cells and ECM, neoinnervation, altered mechanical signaling, and expression of pro-inflammatory cytokines and ion channels. This review aims to discuss what is known about the contributions of acidity to DDD pain, identify the knowledge gaps on this topic, and propose what research can be conducted to fill these gaps. We must better understand the underlying mechanisms of DDD and the interaction between hyperacidity and nociception to develop better therapeutics for this disease.
Collapse
Affiliation(s)
| | - Joshua D. Stover
- Department of Biomedical EngineeringUniversity of UtahSalt Lake CityUtahUSA
- Department of Oral and Craniofacial SciencesUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Alejandro Almarza
- Department of Oral and Craniofacial SciencesUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Robert D. Bowles
- Department of Biomedical EngineeringUniversity of UtahSalt Lake CityUtahUSA
- Department of OrthopaedicsUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
4
|
Rudnik‐Jansen I, van Kruining Kodele S, Creemers L, Joosten B. Biomolecular therapies for chronic discogenic low back pain: A narrative review. JOR Spine 2024; 7:e1345. [PMID: 39114580 PMCID: PMC11303450 DOI: 10.1002/jsp2.1345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/01/2024] [Accepted: 05/01/2024] [Indexed: 08/10/2024] Open
Abstract
Chronic low back pain caused by intervertebral disc (IVD) degeneration, also termed chronic discogenic low back pain (CD-LBP), is one of the most prevalent musculoskeletal diseases. Degenerative processes in the IVD, such as inflammation and extra-cellular matrix breakdown, result in neurotrophin release. Local elevated neurotrophin levels will stimulate sprouting and innervation of sensory neurons. Furthermore, sprouted sensory nerves that are directly connected to adjacent dorsal root ganglia have shown to increase microglia activation, contributing to the maintenance and chronification of pain. Current pain treatments have shown to be insufficient or inadequate for long-term usage. Furthermore, most therapeutic approaches aimed to target the underlying pathogenesis of disc degeneration focus on repair and regeneration and neglect chronic pain. How biomolecular therapies influence the degenerative IVD environment, pain signaling cascades, and innervation and excitability of the sensory neurons often remains unclear. This review addresses the relatively underexplored area of chronic pain treatment for CD-LBP and summarizes effects of therapies aimed for CD-LBP with special emphasis on chronic pain. Approaches based on blocking pro-inflammatory mediators or neurotrophin activity have been shown to hamper neuronal ingrowth into the disc. Furthermore, the tissue regenerative and neuro inhibitory properties of extracellular matrix components or transplanted mesenchymal stem cells are potentially interesting biomolecular approaches to not only block IVD degeneration but also impede pain sensitization. At present, most biomolecular therapies are based on acute IVD degeneration models and thus do not reflect the real clinical chronic pain situation in CD-LBP patients. Future studies should aim at investigating the effects of therapeutic interventions applied in chronic degenerated discs containing established sensory nerve ingrowth. The in-depth understanding of the ramifications from biomolecular therapies on pain (chronification) pathways and pain relief in CD-LBP could help narrow the gap between the pre-clinical bench and clinical bedside for novel CD-LBP therapeutics and optimize pain treatment.
Collapse
Affiliation(s)
- Imke Rudnik‐Jansen
- Department of Anesthesiology and Pain ManagementMaastricht University Medical Center (MUMC+)Maastrichtthe Netherlands
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHeNs)University of MaastrichtMaastrichtthe Netherlands
| | - Sanda van Kruining Kodele
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHeNs)University of MaastrichtMaastrichtthe Netherlands
| | - Laura Creemers
- Department of OrthopedicsUniversity Medical Center UtrechtUtrechtthe Netherlands
| | - Bert Joosten
- Department of Anesthesiology and Pain ManagementMaastricht University Medical Center (MUMC+)Maastrichtthe Netherlands
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHeNs)University of MaastrichtMaastrichtthe Netherlands
| |
Collapse
|
5
|
Tang SN, Salazar-Puerta AI, Heimann MK, Kuchynsky K, Rincon-Benavides MA, Kordowski M, Gunsch G, Bodine L, Diop K, Gantt C, Khan S, Bratasz A, Kokiko-Cochran O, Fitzgerald J, Laudier DM, Hoyland JA, Walter BA, Higuita-Castro N, Purmessur D. Engineered extracellular vesicle-based gene therapy for the treatment of discogenic back pain. Biomaterials 2024; 308:122562. [PMID: 38583365 PMCID: PMC11164054 DOI: 10.1016/j.biomaterials.2024.122562] [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: 08/22/2023] [Revised: 02/23/2024] [Accepted: 03/31/2024] [Indexed: 04/09/2024]
Abstract
Painful musculoskeletal disorders such as intervertebral disc (IVD) degeneration associated with chronic low back pain (termed "Discogenic back pain", DBP), are a significant socio-economic burden worldwide and contribute to the growing opioid crisis. Yet there are very few if any successful interventions that can restore the tissue's structure and function while also addressing the symptomatic pain. Here we have developed a novel non-viral gene therapy, using engineered extracellular vesicles (eEVs) to deliver the developmental transcription factor FOXF1 to the degenerated IVD in an in vivo model. Injured IVDs treated with eEVs loaded with FOXF1 demonstrated robust sex-specific reductions in pain behaviors compared to control groups. Furthermore, significant restoration of IVD structure and function in animals treated with FOXF1 eEVs were observed, with significant increases in disc height, tissue hydration, proteoglycan content, and mechanical properties. This is the first study to successfully restore tissue function while modulating pain behaviors in an animal model of DBP using eEV-based non-viral delivery of transcription factor genes. Such a strategy can be readily translated to other painful musculoskeletal disorders.
Collapse
Affiliation(s)
- Shirley N Tang
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, USA
| | - Ana I Salazar-Puerta
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, USA
| | - Mary K Heimann
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, USA
| | - Kyle Kuchynsky
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, USA
| | | | - Mia Kordowski
- Biophysics Graduate Program, The Ohio State University, USA
| | - Gilian Gunsch
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, USA
| | - Lucy Bodine
- Department of Mechanical Engineering, College of Engineering, The Ohio State University, USA
| | - Khady Diop
- Department of Biology, College of Arts and Sciences, The Ohio State University, USA
| | - Connor Gantt
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, USA
| | - Safdar Khan
- Department of Orthopedics, The Ohio State University Wexner Medical Center, USA
| | - Anna Bratasz
- Small Animal Imaging Center Shared Resources, Wexner Medical Center, USA
| | - Olga Kokiko-Cochran
- Department of Neuroscience, The Ohio State University, USA; Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, USA
| | - Julie Fitzgerald
- Department of Neuroscience, The Ohio State University, USA; Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, USA
| | - Damien M Laudier
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, USA
| | - Judith A Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, The University of Manchester, Manchester, UK; NIHR Manchester Musculoskeletal Biomedical Research Centre, Manchester University, NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Benjamin A Walter
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, USA; Department of Orthopedics, The Ohio State University Wexner Medical Center, USA
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, USA; Biophysics Graduate Program, The Ohio State University, USA; Department of Neurosurgery, The Ohio State University, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, USA.
| | - Devina Purmessur
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, USA; Department of Orthopedics, The Ohio State University Wexner Medical Center, USA.
| |
Collapse
|
6
|
Gansau J, Grossi E, Rodriguez L, Wang M, Laudier DM, Chaudhary S, Hecht AC, Fu W, Sebra R, Liu C, Iatridis JC. TNFR1-mediated senescence and lack of TNFR2-signaling limit human intervertebral disc cell repair in back pain conditions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.22.581620. [PMID: 38948728 PMCID: PMC11212922 DOI: 10.1101/2024.02.22.581620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Poor intervertebral disc (IVD) healing causes IVD degeneration (IVDD) and progression to herniation and back pain. This study identified distinct roles of TNFα-receptors (TNFRs) in contributing to poor healing in painful IVDD. We first isolated IVDD tissue of back pain subjects and determined the complex pro-inflammatory mixture contained many chemokines for recruiting inflammatory cells. Single-cell RNA-sequencing of human IVDD tissues revealed these pro-inflammatory cytokines were dominantly expressed by a small macrophage-population. Human annulus fibrosus (hAF) cells treated with IVDD-conditioned media (CM) underwent senescence with greatly reduced metabolic rates and limited inflammatory responses. TNFR1 inhibition partially restored hAF cell metabolism sufficiently to enable a robust chemokine and cytokine response to CM. We showed that the pro-reparative TNFR2 was very limited on hIVD cell membranes so that TNFR2 inhibition with blocking antibodies or activation using Atsttrin had no effect on hAF cells with CM challenge. However, TNFR2 was expressed in high levels on macrophages identified in scRNA-seq analyses, suggesting their role in repair responses. Results therefore point to therapeutic strategies for painful IVDD involving immunomodulation of TNFR1 signaling in IVD cells to enhance metabolism and enable a more robust inflammatory response including recruitment or delivery of TNFR2 expressing immune cells to enhance IVD repair.
Collapse
Affiliation(s)
- Jennifer Gansau
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Elena Grossi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Levon Rodriguez
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Minghui Wang
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Damien M. Laudier
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Saad Chaudhary
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Andrew C. Hecht
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Wenyu Fu
- Department of Orthopaedics & Rehabilitation, Yale University School of Medicine; New Haven, CT 06510, USA
| | - Robert Sebra
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Chuanju Liu
- Department of Orthopaedics & Rehabilitation, Yale University School of Medicine; New Haven, CT 06510, USA
| | - James C. Iatridis
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| |
Collapse
|
7
|
Song C, Hu P, Peng R, Li F, Fang Z, Xu Y. Bioenergetic dysfunction in the pathogenesis of intervertebral disc degeneration. Pharmacol Res 2024; 202:107119. [PMID: 38417775 DOI: 10.1016/j.phrs.2024.107119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/16/2024] [Accepted: 02/24/2024] [Indexed: 03/01/2024]
Abstract
Intervertebral disc (IVD) degeneration is a frequent cause of low back pain and is the most common cause of disability. Treatments for symptomatic IVD degeneration, including conservative treatments such as analgesics, physical therapy, anti-inflammatories and surgeries, are aimed at alleviating neurological symptoms. However, there are no effective treatments to prevent or delay IVD degeneration. Previous studies have identified risk factors for IVD degeneration such as aging, inflammation, genetic factors, mechanical overload, nutrient deprivation and smoking, but metabolic dysfunction has not been highlighted. IVDs are the largest avascular structures in the human body and determine the hypoxic and glycolytic features of nucleus pulposus (NP) cells. Accumulating evidence has demonstrated that intracellular metabolic dysfunction is associated with IVD degeneration, but a comprehensive review is lacking. Here, by reviewing the physiological features of IVDs, pathological processes and metabolic changes associated with IVD degeneration and the functions of metabolic genes in IVDs, we highlight that glycolytic pathway and intact mitochondrial function are essential for IVD homeostasis. In degenerated NPs, glycolysis and mitochondrial function are downregulated. Boosting glycolysis such as HIF1α overexpression protects against IVD degeneration. Moreover, the correlations between metabolic diseases such as diabetes, obesity and IVD degeneration and their underlying molecular mechanisms are discussed. Hyperglycemia in diabetic diseases leads to cell senescence, the senescence-associated phenotype (SASP), apoptosis and catabolism of extracellualr matrix in IVDs. Correcting the global metabolic disorders such as insulin or GLP-1 receptor agonist administration is beneficial for diabetes associated IVD degeneration. Overall, we summarized the recent progress of investigations on metabolic contributions to IVD degeneration and provide a new perspective that correcting metabolic dysfunction may be beneficial for treating IVD degeneration.
Collapse
Affiliation(s)
- Chao Song
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Peixuan Hu
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Renpeng Peng
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Feng Li
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| | - Zhong Fang
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| | - Yong Xu
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
8
|
Mahmoud M, Kokozidou M, Gögele C, Werner C, Auffarth A, Kohl B, Mrosewski I, Schulze-Tanzil GG. Does Vitamin K2 Influence the Interplay between Diabetes Mellitus and Intervertebral Disc Degeneration in a Rat Model? Nutrients 2023; 15:2872. [PMID: 37447201 DOI: 10.3390/nu15132872] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Intervertebral disc (IVD) degeneration is a common cause of low back pain in diabetes mellitus type 2 (T2DM) patients. Its pathogenesis and the vitamin (vit.) K2 influence on this disease remain unclear. Lumbar motion segments of male Zucker Diabetes Fatty (ZDF) rats (non-diabetic [control] and diabetic; fed without or with vit. K2) were used. Femur lengths and vertebral epiphyseal cross-section areas were measured. IVDs were histopathologically examined. Protein synthesis and gene expression of isolated IVD fibrochondrocytes were analyzed. T2DM rats showed histopathological IVD degeneration. Femur lengths and epiphyseal areas were smaller in T2DM rats regardless of vit. K2 feeding. Fibrochondrocytes synthesized interleukin (IL)-24 and IL-10 with no major differences between groups. Alpha smooth muscle actin (αSMA) was strongly expressed, especially in cells of vit. K2-treated animals. Gene expression of aggrecan was low, and that of collagen type 2 was high in IVD cells of diabetic animals, whether treated with vit. K2 or not. Suppressor of cytokine signaling (Socs)3 and heme oxygenase (Hmox)1 gene expression was highest in the cells of diabetic animals treated with vit. K2. Vit. K2 influenced the expression of some stress-associated markers in IVD cells of diabetic rats, but not that of IL-10 and IL-24.
Collapse
Affiliation(s)
- Mohamed Mahmoud
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Maria Kokozidou
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Clemens Gögele
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Christian Werner
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Alexander Auffarth
- Department of Orthopedics and Traumatology, Paracelsus Medical University, Müllner-Hauptstraße 48, 5020 Salzburg, Austria
| | - Benjamin Kohl
- Department of Traumatology and Reconstructive Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Ingo Mrosewski
- MVZ MDI Limbach Berlin, Aroser Alle 84, 13407 Berlin, Germany
| | - Gundula Gesine Schulze-Tanzil
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| |
Collapse
|
9
|
Gallate ZS, D'Erminio DN, Nasser P, Laudier DM, Iatridis JC. Galectin-3 and RAGE differentially control advanced glycation endproduct-induced collagen damage in murine intervertebral disc organ culture. JOR Spine 2023; 6:e1254. [PMID: 37361328 PMCID: PMC10285763 DOI: 10.1002/jsp2.1254] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/08/2023] [Accepted: 03/07/2023] [Indexed: 06/28/2023] Open
Abstract
Background Back and neck pain are leading causes of global disability that are associated with intervertebral disc (IVD) degeneration. Causes of IVD degeneration are multifactorial, and diet, age, and diabetes have all been linked to IVD degeneration. Advanced glycation endproducts (AGEs) accumulate in the IVD as a result of aging, diet, and diabetes, and AGE accumulation in the IVD has been shown to induce oxidative stress and catabolic activity that result in collagen damage. An association between AGE accumulation and IVD degeneration is emerging, yet mechanism behind this association remains unclear. The Receptor for AGEs (RAGE) is thought to induce catabolic responses in the IVD, and the AGE receptor Galectin 3 (Gal3) had a protective effect in other tissue systems but has not been evaluated in the IVD. Methods This study used an IVD organ culture model with genetically modified mice to analyze the roles of RAGE and Gal3 in an AGE challenge. Results Gal3 was protective against an AGE challenge in the murine IVD ex vivo, limiting collagen damage and biomechanical property changes. Gal3 receptor levels in the AF significantly decreased upon an AGE challenge. RAGE was necessary for AGE-induced collagen damage in the IVD, and RAGE receptor levels in the AF significantly increased upon AGE challenge. Discussion These findings suggest both RAGE and Gal3 are important in the IVD response to AGEs and highlight Gal3 as an important receptor with protective effects on collagen damage. This research improves understanding the mechanisms of AGE-induced IVD degeneration and suggests Gal3 receptor modulation as a potential target for preventative and therapeutic treatment for IVD degeneration.
Collapse
Affiliation(s)
- Zachary S. Gallate
- Leni & Peter W. May Department of OrthopedicsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Danielle N. D'Erminio
- Leni & Peter W. May Department of OrthopedicsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Philip Nasser
- Leni & Peter W. May Department of OrthopedicsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Damien M. Laudier
- Leni & Peter W. May Department of OrthopedicsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - James C. Iatridis
- Leni & Peter W. May Department of OrthopedicsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
10
|
Foreman M, Maddy K, Patel A, Reddy A, Costello M, Lucke-Wold B. Differentiating Lumbar Spinal Etiology from Peripheral Plexopathies. Biomedicines 2023; 11:756. [PMID: 36979737 PMCID: PMC10044821 DOI: 10.3390/biomedicines11030756] [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: 02/04/2023] [Revised: 02/20/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Clinicians have managed and treated lower back pain since the earliest days of practice. Historically, lower back pain and its accompanying symptoms of radiating leg pain and muscle weakness have been recognized to be due to any of the various lumbar spine pathologies that lead to the compression of the lumbar nerves at the root, the most common of which is the radiculopathy known as sciatica. More recently, however, with the increased rise in chronic diseases, the importance of differentially diagnosing a similarly presenting pathology, known as lumbosacral plexopathy, cannot be understated. Given the similar clinical presentation of lumbar spine pathologies and lumbosacral plexopathies, it can be difficult to differentiate these two diagnoses in the clinical setting. Resultingly, the inappropriate diagnosis of either pathology can result in ineffective clinical management. Thus, this review aims to aid in the clinical differentiation between lumbar spine pathology and lumbosacral plexopathy. Specifically, this paper delves into spine and plexus anatomy, delineates the clinical assessment of both pathologies, and highlights powerful diagnostic tools in the hopes of bolstering appropriate diagnosis and treatment. Lastly, this review will describe emerging treatment options for both pathologies in the preclinical and clinical realms, with a special emphasis on regenerative nerve therapies.
Collapse
Affiliation(s)
- Marco Foreman
- Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA
| | - Krisna Maddy
- Department of Neurosurgery, University of Miami, Miami, FL 33136, USA
| | - Aashay Patel
- Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA
| | - Akshay Reddy
- Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA
| | - Meredith Costello
- Department of Neurosurgery, University of Miami, Miami, FL 33136, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
11
|
Savadipour A, Palmer D, Ely EV, Collins KH, Garcia-Castorena JM, Harissa Z, Kim YS, Oestrich A, Qu F, Rashidi N, Guilak F. The role of PIEZO ion channels in the musculoskeletal system. Am J Physiol Cell Physiol 2023; 324:C728-C740. [PMID: 36717101 PMCID: PMC10027092 DOI: 10.1152/ajpcell.00544.2022] [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: 12/06/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 02/01/2023]
Abstract
PIEZO1 and PIEZO2 are mechanosensitive cation channels that are highly expressed in numerous tissues throughout the body and exhibit diverse, cell-specific functions in multiple organ systems. Within the musculoskeletal system, PIEZO1 functions to maintain muscle and bone mass, sense tendon stretch, and regulate senescence and apoptosis in response to mechanical stimuli within cartilage and the intervertebral disc. PIEZO2 is essential for transducing pain and touch sensations as well as proprioception in the nervous system, which can affect musculoskeletal health. PIEZO1 and PIEZO2 have been shown to act both independently as well as synergistically in different cell types. Conditions that alter PIEZO channel mechanosensitivity, such as inflammation or genetic mutations, can have drastic effects on these functions. For this reason, therapeutic approaches for PIEZO-related disease focus on altering PIEZO1 and/or PIEZO2 activity in a controlled manner, either through inhibition with small molecules, or through dietary control and supplementation to maintain a healthy cell membrane composition. Although many opportunities to better understand PIEZO1 and PIEZO2 remain, the studies summarized in this review highlight how crucial PIEZO channels are to musculoskeletal health and point to promising possible avenues for their modulation as a therapeutic target.
Collapse
Affiliation(s)
- Alireza Savadipour
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| | - Daniel Palmer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Erica V Ely
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Kelsey H Collins
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Jaquelin M Garcia-Castorena
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Zainab Harissa
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Yu Seon Kim
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Arin Oestrich
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Feini Qu
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Neda Rashidi
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| |
Collapse
|
12
|
Role of Advanced Glycation End Products in Intervertebral Disc Degeneration: Mechanism and Therapeutic Potential. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7299005. [PMID: 36573114 PMCID: PMC9789911 DOI: 10.1155/2022/7299005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/22/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022]
Abstract
The incidence of low back pain caused by lumbar disc degeneration is high, and it can lead to loss of work ability and impose heavy social and economic burdens. The pathogenesis of low back pain is unclear, and there are no effective treatments. With age, the deposition of advanced glycation end products (AGEs) in intervertebral disc (IVD) gradually increases and is accelerated by diabetes and a high-AGEs diet, leading to destruction of the annulus fibrosus (AF), nucleus pulposus (NP), and cartilage endplate (CEP) and finally intervertebral disc degeneration (IDD). Reducing the accumulation of AGEs in IVD and blocking the transmission of downstream signals caused by AGEs have a significant effect on alleviating IDD. In this review, we summarize the mechanism by which AGEs induce IDD and potential treatment strategies.
Collapse
|
13
|
Krull CM, Rife J, Klamer B, Purmessur D, Walter BA. Pericellular heparan sulfate proteoglycans: Role in regulating the biosynthetic response of nucleus pulposus cells to osmotic loading. JOR Spine 2022; 5:e1209. [PMID: 35783912 PMCID: PMC9238280 DOI: 10.1002/jsp2.1209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/26/2022] [Accepted: 05/12/2022] [Indexed: 12/04/2022] Open
Abstract
Background Daily physiologic loading causes fluctuations in hydration of the intervertebral disc (IVD); thus, the embedded cells experience cyclic alterations to their osmotic environment. These osmotic fluctuations have been described as a mechanism linking mechanics and biology, and have previously been shown to promote biosynthesis in chondrocytes. However, this phenomenon has yet to be fully interrogated in the IVD. Additionally, the specialized extracellular matrix surrounding the cells, the pericellular matrix (PCM), transduces the biophysical signals that cells ultimately experience. While it is known that the PCM is altered in disc degeneration, whether it disrupts normal osmotic mechanotransduction has yet to be determined. Thus, our objectives were to assess: (1) whether dynamic osmotic conditions stimulate biosynthesis in nucleus pulposus cells, and (2) whether pericellular heparan sulfate proteoglycans (HSPGs) modulate the biosynthetic response to osmotic loading. Methods Bovine nucleus pulposus cells isolated with retained PCM were encapsulated in 1.5% alginate beads and treated with or without heparinase III, an enzyme that degrades the pericellular HSPGs. Beads were subjected to 1 h of daily iso-osmotic, hyper-osmotic, or hypo-osmotic loading for 1, 2, or 4 weeks. At each timepoint the total amount of extracellular and pericellular sGAG/DNA were quantified. Additionally, whether osmotic loading triggered alterations to HSPG sulfation was assessed via immunohistochemistry for the heparan sulfate 6-O-sulfertransferase 1 (HS6ST1) enzyme. Results Osmotic loading significantly influenced sGAG/DNA accumulation with a hyper-osmotic change promoting the greatest sGAG/DNA accumulation in the pericellular region compared with iso-osmotic conditions. Heparanase-III treatment significantly reduced extracellular sGAG/DNA but pericellular sGAG was not affected. HS6ST1 expression was not affected by osmotic loading. Conclusion Results suggest that hyper-osmotic loading promotes matrix synthesis and that modifications to HSPGs directly influence the metabolic responses of cells to osmotic fluctuations. Collectively, results suggest degeneration-associated modifications to pericellular HSPGs may contribute to the altered mechanobiology observed in disease.
Collapse
Affiliation(s)
- Carly M. Krull
- Department of Biomedical EngineeringThe Ohio State UniversityColumbusOhioUSA
| | - Jordan Rife
- Department of Biomedical EngineeringThe Ohio State UniversityColumbusOhioUSA
| | - Brett Klamer
- Department of Biomedical Informatics, Center for BiostatisticsThe Ohio State UniversityColumbusOhioUSA
| | - Devina Purmessur
- Department of Biomedical EngineeringThe Ohio State UniversityColumbusOhioUSA
- Department of OrthopedicsThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Spine Research InstituteThe Ohio State UniversityColumbusOhioUSA
| | - Benjamin A. Walter
- Department of Biomedical EngineeringThe Ohio State UniversityColumbusOhioUSA
- Department of OrthopedicsThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Spine Research InstituteThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
14
|
Li C, Chen J, Lv Y, Liu Y, Guo Q, Wang J, Wang C, Hu P, Liu Y. Recent Progress in Electrospun Nanofiber-Based Degenerated Intervertebral Disc Repair. ACS Biomater Sci Eng 2021; 8:16-31. [PMID: 34913688 DOI: 10.1021/acsbiomaterials.1c00970] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Annulus fibrosus fissure and fibrosis of nucleus pulposus are severe morphological characteristics of intervertebral disc degeneration. Currently, surgery or drugs are used to relieve pain in such cases. Tissue engineering is a new multidisciplinary strategy with great potential for use in joint replacement and organ regeneration. Based on the natural anatomy of intervertebral discs, intervertebral disc scaffolds are fabricated by exploiting the special arrangement of extracellular matrix fibers. Electrospun nanofibers possess clear advantages in repairing degenerated intervertebral discs. This article reviews and summarizes recently developed methods for improving and fabricating electrospun nanofiber annulus fibrosus scaffolds in terms of nanofiber alignment, material selection, loading additives, and the progress made in combining other advanced technologies with electrospun nanofibers. In addition, the improvement in mechanical properties and biocompatibility of nucleus pulposus scaffolds by electrospun nanofiber-reinforced hydrogels is discussed. Finally, complete intervertebral disc scaffolds can be fabricated using the disc-like angle-ply structure and other emerging fabrication methods. Taken together, electrospun nanofiber intervertebral disc scaffolds are promising for clinical applications.
Collapse
Affiliation(s)
- Chenxi Li
- Beijing Key Laboratory of Advanced Functional Polymer Composites, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jia Chen
- Beijing Key Laboratory of Advanced Functional Polymer Composites, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yarong Lv
- Beijing Key Laboratory of Advanced Functional Polymer Composites, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yueqi Liu
- Beijing Key Laboratory of Advanced Functional Polymer Composites, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Quanyi Guo
- Institute of Orthopedics, the Fourth Medical Center, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Jiandong Wang
- Division of Breast Surgery, Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Ce Wang
- Alan G. MacDiarmid Institute, Jilin University, Changchun, Jilin 130012, China
| | - Ping Hu
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Yong Liu
- Beijing Key Laboratory of Advanced Functional Polymer Composites, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
15
|
Di Pauli von Treuheim T, Torre OM, Ferreri ED, Nasser P, Abbondandolo A, Delgado Caceres M, Lin D, Docheva D, Iatridis JC. Tenomodulin and Chondromodulin-1 Are Both Required to Maintain Biomechanical Function and Prevent Intervertebral Disc Degeneration. Cartilage 2021; 13:604S-614S. [PMID: 34486420 PMCID: PMC8804743 DOI: 10.1177/19476035211029696] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE The underlying mechanisms and molecular factors influencing intervertebral disc (IVD) homeostasis and degeneration remain clinically relevant. Tenomodulin (Tnmd) and chondromodulin (Chm1) are antiangiogenic transmembrane glycoproteins, with cleavable C-terminus, expressed by IVD cells that are implicated in the onset of degenerative processes. We evaluate the organ-level biomechanical impact of knocking out Tnmd alone, and Tnmd and Chm1, simultaneously. DESIGN Caudal (c5-8) and lumbar vertebrae (L1-4) of skeletally mature male and female 9-month-old wildtype (WT), Tnmd knockout (Tnmd-/-), and Tnmd/Chm1 double knockout (Tnmd-/-/Chm-/-) mice were used (n = 9-13 per group). Disc height index (DHI), histomorphological changes, and axial, torsional, creep, and failure biomechanical properties were evaluated. Differences were assessed by one-way ANOVA with post hoc Bonferroni-corrected comparisons (P < 0.05). RESULTS Tnmd-/-/Chm1-/- IVDs displayed increased DHI and histomorphological scores that indicated increased IVD degeneration compared to the WT and Tnmd-/- groups. Double knockout IVDs required significantly less torque and energy to initiate torsional failure. Creep parameters were comparable between all groups, except for the slow time constant, which indicated faster outward fluid flow. Tnmd-/- IVDs lost fluid faster than the WT group, and this effect was amplified in the double knockout IVDs. CONCLUSION Knocking out Tnmd and Chm1 affects IVD fluid flow and organ-level biomechanical function and therefore may play a role in contributing to IVD degeneration. Larger effects of the Tnmd and Chm1 double knockout mice compared to the Tnmd single mutant suggest that Chm1 may play a compensatory role in the Tnmd single mutant IVDs.
Collapse
Affiliation(s)
| | - Olivia M. Torre
- Leni & Peter W. May Department of
Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily D. Ferreri
- Leni & Peter W. May Department of
Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Philip Nasser
- Leni & Peter W. May Department of
Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Angelica Abbondandolo
- Leni & Peter W. May Department of
Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manuel Delgado Caceres
- Experimental Trauma Surgery, Department
of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Dasheng Lin
- Orthopaedic Center of People’s
Liberation Army, The Affiliated Southeast Hospital of Xiamen University, Zhangzhou,
China
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department
of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - James C. Iatridis
- Leni & Peter W. May Department of
Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA,James C. Iatridis, Leni & Peter W. May
Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, One Gustave
Levy Place, Box 1188, New York, NY 10029-6574, USA.
| |
Collapse
|
16
|
Lakstins K, Arnold L, Gunsch G, Flanigan D, Khan S, Gadde N, Jones B, Agarwal G, Purmessur D. Characterization of the human intervertebral disc cartilage endplate at the molecular, cell, and tissue levels. J Orthop Res 2021; 39:1898-1907. [PMID: 32915471 DOI: 10.1002/jor.24854] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/29/2020] [Accepted: 09/08/2020] [Indexed: 02/04/2023]
Abstract
Given the importance of the cartilage endplate (CEP) in low back pain (LBP), there is a need to characterize the human CEP at the molecular, cell, and tissue levels to inform treatment strategies that target it. The goal of this study was to characterize the structure, matrix composition, and cell phenotype of the human CEP compared with adjacent tissues within the intervertebral joint: the nucleus pulposus (NP), annulus fibrosus (AF), and articular cartilage (AC). Isolated CEP, NP, AF, and AC tissues and cells were evaluated for cell morphology, matrix composition, collagen structure, glycosaminoglycan content, and gene and protein expression. The CEP contained elongated cells that mainly produce a collagen-rich interterritorial matrix and a proteoglycan-rich territorial matrix. The CEP contained significantly fewer glycosaminoglycans than the NP tissue. Significant differences in matrix and cell marker gene expression were observed between CEP and NP or AF, with the greatest differences between CEP and AC. We were able to distinguish NP from CEP cells using collagen-10 (COLX), highlighting COLX as a potential CEP marker. Our findings suggest that at the cell and tissue levels, the CEP demonstrates both similarities and differences when compared with NP, AF, and hyaline AC. This study highlights a unique structure, matrix composition, and cell phenotype for the human CEP and can help to inform regenerative strategies that target the intervertebral disc joint in chronic LBP.
Collapse
Affiliation(s)
- Katherine Lakstins
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Lauren Arnold
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Gilian Gunsch
- Department of Biology, The Ohio State University, Columbus, Ohio, USA
| | - David Flanigan
- Department of Orthopaedics, The Ohio State University, Columbus, Ohio, USA
| | - Safdar Khan
- Department of Orthopaedics, The Ohio State University, Columbus, Ohio, USA
| | - Nikhit Gadde
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Blain Jones
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Gunjan Agarwal
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Devina Purmessur
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.,Department of Orthopaedics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
17
|
Ekram S, Khalid S, Bashir I, Salim A, Khan I. Human umbilical cord-derived mesenchymal stem cells and their chondroprogenitor derivatives reduced pain and inflammation signaling and promote regeneration in a rat intervertebral disc degeneration model. Mol Cell Biochem 2021; 476:3191-3205. [PMID: 33864569 DOI: 10.1007/s11010-021-04155-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 04/02/2021] [Indexed: 12/21/2022]
Abstract
Intervertebral disc (IVD) degeneration is an asymptomatic pathophysiological condition and a strong causative factor of low back pain. There is no cure available except spinal fusion and pain management. Stem cell-based regenerative medicine is being considered as an alternative approach to treat disc diseases. The current study aimed to differentiate human umbilical cord-mesenchymal stem cells (hUC-MSCs) into chondrocyte-like cells and to elucidate their feasibility and efficacy in the degenerated IVD rat model. Chondrogenic induction medium was used to differentiate hUC-MSCs into chondroprogenitors. Rat tail IVD model was established with three consecutive coccygeal discs. qPCR was performed to quantify the molecular markers of pain and inflammation. Histological staining was performed to evaluate the degree of regeneration. Induced chondroprogenitors showed the expression of chondrogenic genes, SOX9, TGF-β1, ACAN, BMP2, and GDF5. Immunocytochemical staining showed positive expression of chondrogenic proteins SOX9, TGF-β1, TGF-β2, and Collagen 2. In in vivo study, transplanted chondroprogenitors showed better survival, homing, and distribution in IVD as compared to normal MSCs. Expression of pain and inflammatory genes at day 5 of cell transplantation modulated immune response significantly. The transplanted labeled MSCs and induced chondroprogenitors differentiated into functional nucleus pulposus (NP) cells as evident from co-localization of red (DiI) and green fluorescence for SOX9, TGF-β1, and TGF-β2. Alcian blue and H & E staining showed standard histological features, indicating better preservation of the NP structure and cellularity than degenerated discs. hUC-MSCs-derived chondroprogenitors showed better regeneration potential as compared to normal MSCs. The pain and inflammation genes were downregulated in the treated group as compared to the degenerated IVD.
Collapse
Affiliation(s)
- Sobia Ekram
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Shumaila Khalid
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Imtiaz Bashir
- Zainab Panjwani Memorial Hospital, Mohammadali Habib Road, Numaish Karachi, 74800, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
18
|
Le Maitre CL, Dahia CL, Giers M, Illien‐Junger S, Cicione C, Samartzis D, Vadala G, Fields A, Lotz J. Development of a standardized histopathology scoring system for human intervertebral disc degeneration: an Orthopaedic Research Society Spine Section Initiative. JOR Spine 2021; 4:e1167. [PMID: 34337340 PMCID: PMC8313169 DOI: 10.1002/jsp2.1167] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/25/2021] [Accepted: 06/07/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Histopathological analysis of intervertebral disc (IVD) tissues is a critical domain of back pain research. Identification, description, and classification of attributes that distinguish abnormal tissues form a basis for probing disease mechanisms and conceiving novel therapies. Unfortunately, lack of standardized methods and nomenclature can limit comparisons of results across studies and prevent organizing information into a clear representation of the hierarchical, spatial, and temporal patterns of IVD degeneration. Thus, the following Orthopaedic Research Society (ORS) Spine Section Initiative aimed to develop a standardized histopathology scoring scheme for human IVD degeneration. METHODS Guided by a working group of experts, this prospective process entailed a series of stages that consisted of reviewing and assessing past grading schemes, surveying IVD researchers globally on current practice and recommendations for a new grading system, utilizing expert opinion a taxonomy of histological grading was developed, and validation performed. RESULTS A standardized taxonomy was developed, which showed excellent intra-rater reliability for scoring nucleus pulposus (NP), annulus fibrosus (AF), and cartilaginous end plate (CEP) regions (interclass correlation [ICC] > .89). The ability to reliably detect subtle changes varied by IVD region, being poorest in the NP (ICC: .89-.95) where changes at the cellular level were important, vs the AF (ICC: .93-.98), CEP (ICC: .97-.98), and boney end plate (ICC: .96-.99) where matrix and structural changes varied more dramatically with degeneration. CONCLUSIONS The proposed grading system incorporates more comprehensive descriptions of degenerative features for all the IVD sub-tissues than prior criteria. While there was excellent reliability, our results reinforce the need for improved training, particularly for novice raters. Future evaluation of the proposed system in real-world settings (eg, at the microscope) will be needed to further refine criteria and more fully evaluate utility. This improved taxonomy could aid in the understanding of IVD degeneration phenotypes and their association with back pain.
Collapse
Affiliation(s)
| | - Chitra L. Dahia
- Orthopaedic Soft Tissue Research ProgramHospital for Special SurgeryNew YorkNew YorkUSA
- Department of Cell and Developmental BiologyWeill Cornell Medicine, Graduate School of Medical SciencesNew YorkNew YorkUSA
| | - Morgan Giers
- School of Chemical, Biological, and Environmental EngineeringOregon State UniversityCorvallisOregonUSA
| | | | - Claudia Cicione
- Laboratory of Regenerative Orthopaedics, Department of Orthopaedic and Trauma SurgeryCampus Bio‐Medico University of RomeRomeItaly
| | - Dino Samartzis
- Department of Orthopaedic SurgeryRush University Medical CenterChicagoIllinoisUSA
- International Spine Research and Innovation InitiativeRush University Medical CenterChicagoIllinoisUSA
| | - Gianluca Vadala
- Laboratory of Regenerative Orthopaedics, Department of Orthopaedic and Trauma SurgeryCampus Bio‐Medico University of RomeRomeItaly
| | - Aaron Fields
- Department of Orthopaedic SurgeryUniversity of California at San FranciscoSan FranciscoCaliforniaUSA
| | - Jeffrey Lotz
- Department of Orthopaedic SurgeryUniversity of California at San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
19
|
Lakstins K, Yeater T, Arnold L, Khan S, Hoyland JA, Purmessur D. Investigating the role of culture conditions on hypertrophic differentiation in human cartilage endplate cells. J Orthop Res 2021; 39:1204-1216. [PMID: 32285966 DOI: 10.1002/jor.24692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/18/2020] [Accepted: 03/28/2020] [Indexed: 02/04/2023]
Abstract
Cartilage endplate degeneration/calcification has been linked to the onset and progression of intervertebral disc degeneration and there is a critical need to understand mechanisms, such as hypertrophic differentiation, of cartilage endplate degeneration/calcification to inform treatment strategies for discogenic back pain. In vitro cell culture conditions capable of inducing hypertrophic differentiation are used to study pathophysiological mechanisms in articular chondrocytes, but culture conditions capable of inducing a hypertrophic cartilage endplate cell phenotype have yet to be explored. The goal of this study was to investigate the role of culture conditions capable of inducing hypertrophic differentiation in articular chondrocytes on hypertrophic differentiation in human cartilage endplate cells. Isolated human cartilage endplate cells were cultured as pellets for 21 days at either 5% O2 (physiologic for cartilage) or 20.7% O2 (hyperoxic) and treated with 10% fetal bovine serum or Wnt agonist, two stimuli used to induce hypertrophic differentiation in articular chondrocytes. Cartilage endplate cells did not exhibit a hypertrophic cell morphology in response to fetal bovine serum or Wnt agonist but did display other hallmarks of chondrocyte hypertrophy and degeneration such as hypertrophic gene and protein expression, and a decrease in healthy proteoglycans and an increase in fibrous collagen accumulation. These findings demonstrate that cartilage endplate cells take on a degenerative phenotype in response to hypertrophic stimuli in vitro, but do not undergo classical changes in morphology associated with hypertrophic differentiation regardless of oxygen levels, highlighting potential differences in the response of cartilage endplate cells versus articular chondrocytes to the same stimuli.
Collapse
Affiliation(s)
- Katherine Lakstins
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Taylor Yeater
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Lauren Arnold
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Safdar Khan
- Department of Orthopedics, The Ohio State University, Columbus, Ohio
| | - Judith A Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, The University of Manchester, Manchester, UK.,NIHR Manchester Biomedical Research Centre, Central Manchester Foundation Trust, Manchester Academic Health Science Centre, School of Biological Sciences, Manchester, UK
| | - Devina Purmessur
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio.,Department of Orthopedics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
20
|
Castro APG. Computational Challenges in Tissue Engineering for the Spine. Bioengineering (Basel) 2021; 8:25. [PMID: 33671854 PMCID: PMC7918040 DOI: 10.3390/bioengineering8020025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/04/2021] [Accepted: 02/13/2021] [Indexed: 12/17/2022] Open
Abstract
This paper deals with a brief review of the recent developments in computational modelling applied to innovative treatments of spine diseases. Additionally, it provides a perspective on the research directions expected for the forthcoming years. The spine is composed of distinct and complex tissues that require specific modelling approaches. With the advent of additive manufacturing and increasing computational power, patient-specific treatments have moved from being a research trend to a reality in clinical practice, but there are many issues to be addressed before such approaches become universal. Here, it is identified that the major setback resides in validation of these computational techniques prior to approval by regulatory agencies. Nevertheless, there are very promising indicators in terms of optimised scaffold modelling for both disc arthroplasty and vertebroplasty, powered by a decisive contribution from imaging methods.
Collapse
Affiliation(s)
- André P G Castro
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| |
Collapse
|
21
|
Yang X, Chen Z, Chen C, Han C, Zhou Y, Li X, Tian H, Cheng X, Zhang K, Qin A, Zhou T, Zhao J. Bleomycin induces fibrotic transformation of bone marrow stromal cells to treat height loss of intervertebral disc through the TGFβR1/Smad2/3 pathway. Stem Cell Res Ther 2021; 12:34. [PMID: 33413668 PMCID: PMC7791639 DOI: 10.1186/s13287-020-02093-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/10/2020] [Indexed: 01/07/2023] Open
Abstract
Background Lower back pain is often accredited to loss of intervertebral disc (IVD) height and compromised spine stability as a result of intervertebral disc degeneration (IVDD). We aim to locally use bleomycin to induce the fibrotic transformation of bone marrow stromal cells (BMSCs) as a means to induce reparative fibrosis to slow down the height loss. Methods IVDs from patients were gathered for histological examination. The expression of the transforming growth factor beta 1 (TGF-β) signaling pathway was determined by qPCR and western blotting. Nucleus pulposus (NP) cells, annulus fibrosus (AF) cells, and the rats’ bone marrow stromal cells (BMSC) were cultured and their responsiveness to bleomycin was evaluated by Cell Counting Kit-8, comet assay, transwell migration, and wound healing assays. Rat IVDD models were created by puncture and rescued by bleomycin injection, and the effectiveness was evaluated by images (X-ray and MRI) and atomic force microscope. Results Histological examination showed increased levels of pro-fibrotic markers in IVDD tissues from patients. AF cells and BMSC cells were induced to adopt a pro-fibrotic phenotype with increased expression fibrotic markers Col1a1, Col3a1, and FSP1. The pro-fibrotic effect of bleomycin on AF cells and BMSCs was in part due to the activation of the TGFβ-TGFβR1-SMAD2/3 signaling pathway. Pharmacological inhibition or gene knock-down of TGFβR1 could mitigate the pro-fibrotic effects. Conclusion Locally, injection of bleomycin in rats’ IVD induced rapid fibrosis and maintained its height through the TGFβ-TGFβR1-SMAD2/3 signaling pathway.
Collapse
Affiliation(s)
- Xiao Yang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Zhiqian Chen
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Chen Chen
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Chen Han
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Yifan Zhou
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Xunlin Li
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Haijun Tian
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Xiaofei Cheng
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Kai Zhang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - An Qin
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| | - Tangjun Zhou
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| | - Jie Zhao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
22
|
Lakstins K, Arnold L, Gunsch G, Khan S, Moore S, Purmessur D. Characterization of bovine and canine animal model cartilage endplates and comparison to human cartilage endplate structure, matrix composition, and cell phenotype. JOR Spine 2020; 3:e1116. [PMID: 33392453 PMCID: PMC7770203 DOI: 10.1002/jsp2.1116] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/20/2020] [Accepted: 05/28/2020] [Indexed: 12/19/2022] Open
Abstract
There is a need to further explore mechanisms of cartilage endplate (CEP) degeneration, due to its role in the onset and progression of intervertebral disc degeneration and low back pain. Therefore, the goal of this study was to evaluate structure, matrix composition, and cell phenotype between the human and bovine or canine, both clinically relevant animal models currently used to study the intervertebral disc, CEP. This information may be used in addition to other relevant studies, to help determine optimal animal models for use in studying the role of the CEP in intervertebral disc degeneration and back pain. Endplate structure, matrix composition, cell morphology, and gene expression were evaluated using a picrosirius red/alcian blue and hematoxylin and eosin stain, a dimethylmethylene blue assay, and quantitative reverse transcription polymerase chain reaction. The bovine and canine CEPs were thinner with more rounded cells and thicker bony endplates. The canine CEP contained significantly more sulfated glycosaminoglycans. The bovine CEP demonstrated higher expression of ACAN, COL1, and COL2 and lower expression of T, FBLN1, and collagen X (COLX) compared to the human CEP. The canine CEP had higher COL2 and lower COL1, KRT19, MKX, FBLN1, COLX expression compared to human. These similarities and differences between human and bovine or canine CEP are important to consider when evaluating which animal model is most optimal to use in future studies, interpreting research findings using these animal models and assessing translatability to the human condition.
Collapse
Affiliation(s)
- Katherine Lakstins
- Department of Biomedical EngineeringThe Ohio State UniversityColumbusOhioUSA
| | - Lauren Arnold
- Department of Biomedical EngineeringThe Ohio State UniversityColumbusOhioUSA
| | - Gilian Gunsch
- Department of BiologyThe Ohio State UniversityColumbusOhioUSA
| | - Safdar Khan
- Department of OrthopaedicsThe Ohio State UniversityColumbusOhioUSA
| | - Sarah Moore
- Department of Veterinary Clinical SciencesThe Ohio State UniversityColumbusOhioUSA
| | - Devina Purmessur
- Department of Biomedical EngineeringThe Ohio State UniversityColumbusOhioUSA
- Department of OrthopaedicsThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
23
|
Zeldin L, Mosley GE, Laudier D, Gallate ZS, Gansau J, Hoy RC, Poeran J, Iatridis JC. Spatial mapping of collagen content and structure in human intervertebral disk degeneration. JOR Spine 2020; 3:e1129. [PMID: 33392461 PMCID: PMC7770200 DOI: 10.1002/jsp2.1129] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 10/13/2020] [Accepted: 10/18/2020] [Indexed: 12/11/2022] Open
Abstract
Collagen plays a key structural role in both the annulus fibrosus (AF) and nucleus pulposus (NP) of intervertebral disks (IVDs). Changes in collagen content with degeneration suggest a shift from collagen type II to type I within the NP, and the activation of pro-inflammatory factors is indicative of fibrosis throughout. While IVD degeneration is considered a fibrotic process, an increase in collagen content with degeneration, reflective of fibrosis, has not been demonstrated. Additionally, changes in collagen content and structure in human IVDs with degeneration have not been characterized with high spatial resolution. The collagen content of 23 human lumbar L2/3 or L3/4 IVDs was quantified using second harmonic generation imaging (SHG) and multiple image processing algorithms, and these parameters were correlated with the Rutges histological degeneration grade. In the NP, SHG intensity increased with degeneration grade, suggesting fibrotic collagen deposition. In the AF, the entropy of SHG intensity was reduced with degeneration indicating increased collagen uniformity and suggesting less-organized lamellar structure. Collagen orientation entropy decreased throughout most IVD regions with increasing degeneration grade, further supporting a loss in collagen structural complexity. Overall, SHG imaging enabled visualization and quantification of IVD collagen content and organization with degeneration. There was an observed shift from an initially complex structure to more uniform structure with loss of microstructural elements and increased NP collagen polarity, suggesting fibrotic remodeling.
Collapse
Affiliation(s)
- Lawrence Zeldin
- Leni & Peter W. May Department of OrthopedicsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Grace E. Mosley
- Leni & Peter W. May Department of OrthopedicsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Damien Laudier
- Leni & Peter W. May Department of OrthopedicsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Zachary S. Gallate
- Leni & Peter W. May Department of OrthopedicsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jennifer Gansau
- Leni & Peter W. May Department of OrthopedicsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Robert C. Hoy
- Leni & Peter W. May Department of OrthopedicsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jashvant Poeran
- Leni & Peter W. May Department of OrthopedicsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Population Health Science and PolicyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - James C. Iatridis
- Leni & Peter W. May Department of OrthopedicsIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
24
|
Borem R, Walters J, Madeline A, Madeline L, Gill S, Easley J, Mercuri J. Characterization of chondroitinase-induced lumbar intervertebral disc degeneration in a sheep model intended for assessing biomaterials. J Biomed Mater Res A 2020; 109:1232-1246. [PMID: 33040470 DOI: 10.1002/jbm.a.37117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/04/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023]
Abstract
Intervertebral disc (IVD) degeneration (IVDD) leads to structural and functional changes. Biomaterials for restoring IVD function and promoting regeneration are currently being investigated; however, such approaches require validation using animal models that recapitulate clinical, biochemical, and biomechanical hallmarks of the human pathology. Herein, we comprehensively characterized a sheep model of chondroitinase-ABC (ChABC) induced IVDD. Briefly, ChABC (1 U) was injected into the L1/2 , L2/3 , and L3/4 IVDs. Degeneration was assessed via longitudinal magnetic resonance (MR) and radiographic imaging. Additionally, kinematic, biochemical, and histological analyses were performed on explanted functional spinal units (FSUs). At 17-weeks, ChABC treated IVDs demonstrated significant reductions in MR index (p = 0.030) and disc height (p = 0.009) compared with pre-operative values. Additionally, ChABC treated IVDs exhibited significantly increased creep displacement (p = 0.004) and axial range of motion (p = 0.007) concomitant with significant decreases in tensile (p = 0.034) and torsional (p = 0.021) stiffnesses and long-term viscoelastic properties (p = 0.016). ChABC treated IVDs also exhibited a significant decrease in NP glycosaminoglycan: hydroxyproline ratio (p = 0.002) and changes in microarchitecture, particularly in the NP and endplates, compared with uninjured IVDs. Taken together, this study demonstrated that intradiscal injection of ChABC induces significant degeneration in sheep lumbar IVDs and the potential for using this model in evaluating biomaterials for IVD repair, regeneration, or fusion.
Collapse
Affiliation(s)
- Ryan Borem
- The Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Joshua Walters
- The Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Allison Madeline
- The Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Lee Madeline
- Department of Radiology, Greenville Health System, Greenville, South Carolina, USA
| | - Sanjitpal Gill
- The Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, South Carolina, USA.,Department of Orthopaedic Surgery, Medical Group of the Carolinas-Pelham, Spartanburg Regional Healthcare System, Greer, South Carolina, USA
| | - Jeremiah Easley
- Preclinical Surgical Research Laboratory, Colorado State University, Fort Collins, Colorado, USA
| | - Jeremy Mercuri
- The Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
25
|
Natelson DM, Lai A, Krishnamoorthy D, Hoy RC, Iatridis JC, Illien-Jünger S. Leptin signaling and the intervertebral disc: Sex dependent effects of leptin receptor deficiency and Western diet on the spine in a type 2 diabetes mouse model. PLoS One 2020; 15:e0227527. [PMID: 32374776 PMCID: PMC7202633 DOI: 10.1371/journal.pone.0227527] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
Type 2 diabetes and obesity are associated with back pain in juveniles and adults and are implicated in intervertebral disc (IVD) degeneration. Hypercaloric Western diets are associated with both obesity and type 2 diabetes. The objective of this study was to determine if obesity and type 2 diabetes result in spinal pathology in a sex-specific manner using in vivo diabetic and dietary mouse models. Leptin is an appetite-regulating hormone, and its deficiency leads to polyphagia, resulting in obesity and diabetes. Leptin is also associated with IVD degeneration, and increased expression of its receptor was identified in degenerated IVDs. We used young, leptin receptor deficient (Db/Db) mice to mimic the effect of diet and diabetes on adolescents. Db/Db and Control mice were fed either Western or Control diets, and were sacrificed at 3 months of age. Db/Db mice were obese, while only female mice developed diabetes. Female Db/Db mice displayed altered IVD morphology, with increased intradiscal notochordal band area, suggesting delayed IVD cell proliferation and differentiation, rather than IVD degeneration. Motion segments from Db/Db mice exhibited increased failure risk with decreased torsional failure strength. Db/Db mice also had inferior bone quality, which was most prominent in females. We conclude that obesity and diabetes due to impaired leptin signaling contribute to pathological changes in vertebrae, as well as an immature IVD phenotype, particularly of females, suggesting a sex-dependent role of leptin in the spine.
Collapse
Affiliation(s)
- Devorah M. Natelson
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Alon Lai
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Divya Krishnamoorthy
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Robert C. Hoy
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - James C. Iatridis
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Svenja Illien-Jünger
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, United States of America
- * E-mail:
| |
Collapse
|
26
|
Jiang EY, Sloan SR, Wipplinger C, Kirnaz S, Härtl R, Bonassar LJ. Proteoglycan removal by chondroitinase ABC improves injectable collagen gel adhesion to annulus fibrosus. Acta Biomater 2019; 97:428-436. [PMID: 31425894 DOI: 10.1016/j.actbio.2019.08.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/24/2019] [Accepted: 08/13/2019] [Indexed: 12/14/2022]
Abstract
Intervertebral disc (IVD) herniations are currently treated with interventions that leave the IVD with persistent lesions prone to further herniations. Annulus fibrosus (AF) repair has become of interest as a method to seal defects in the IVD and prevent reherniation, but this requires strong adhesion of the implanted biomaterial to the native AF tissue. Our group has previously developed a high-density collagen (HDC) gel for AF repair and tested its efficacy in vivo, but its adhesion to the AF could be improved. Increased cell adhesion to cartilage has previously been reported through chondroitinase ABC (ChABC) digestion, which removes proteoglycans and increases access to cell binding motifs. Such approaches could also increase biomaterial adhesion to tissue, but the effects of ChABC digestion on AF have yet to be investigated. In this study, ovine AF tissue was digested with either 10 U/mL ChABC or saline for up to 10 min and the effect of this treatment on collagen adhesion between AF tissue samples was investigated by histology and mechanical testing in a lap-shear configuration. ChABC digestion removed proteoglycans within the AF in a time-dependent fashion and enhanced adhesion of the HDC gel to the AF. ChABC digestion increased the elastic toughness and total shear energy of the HDC gel-AF interface by 88% and 46% respectively. ChABC treatment enhanced the adhesion of the HDC gel to the AF without significantly decreasing native AF cell viability. Thus, ChABC digestion is a viable method to improve adhesion of biomaterials for AF repair. STATEMENT OF SIGNIFICANCE: Intervertebral disc herniations are currently treated with interventions that leave persistent lesions in the annulus fibrosus that are prone to further herniations. Annular repair is a promising method to seal lesions and prevent reherniation, but requires strong adhesion of the implanted biomaterial to native annulus fibrosus. Since large proteoglycans like aggrecan occupy regions of the extracellular matrix between collagen fibers in the annulus fibrosus, we hypothesized that removing proteoglycans via chondroitinase digestion would increase the adhesion of annular repair hydrogels. This investigation demonstrated that chondroitinase removed proteoglycans within annulus fibrosus tissue, enhanced the interaction of an injected collagen gel with the native tissue, and mechanically improved adhesion between the collagen gel and annulus fibrosus. This is the first study of its kind to evaluate the biochemical and mechanical effects of short-term chondroitinase digestion on annulus fibrosus tissue.
Collapse
|
27
|
Tendulkar G, Chen T, Ehnert S, Kaps HP, Nüssler AK. Intervertebral Disc Nucleus Repair: Hype or Hope? Int J Mol Sci 2019; 20:3622. [PMID: 31344903 PMCID: PMC6696292 DOI: 10.3390/ijms20153622] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/19/2019] [Accepted: 07/20/2019] [Indexed: 12/28/2022] Open
Abstract
Chronic back pain is a common disability, which is often accredited to intervertebral disc degeneration. Gold standard interventions such as spinal fusion, which are mainly designed to mechanically seal the defect, frequently fail to restore the native biomechanics. Moreover, artificial implants have limited success as a repair strategy, as they do not alter the underlying disease and fail to promote tissue integration and subsequent native biomechanics. The reported high rates of spinal fusion and artificial disc implant failure have pushed intervertebral disc degeneration research in recent years towards repair strategies. Intervertebral disc repair utilizing principles of tissue engineering should theoretically be successful, overcoming the inadequacies of artificial implants. For instance, advances in the development of scaffolds aided with cells and growth factors have opened up new possibilities for repair strategies. However, none has reached the stage of clinical trials in humans. In this review, we describe the hitches encountered in the musculoskeletal field and summarize recent advances in designing tissue-engineered constructs for promoting nucleus pulposus repair. Additionally, the review focuses on the effect of biomaterial aided with cells and growth factors on achieving effective functional reparative potency, highlighting the ways to enhance the efficacy of these treatments.
Collapse
Affiliation(s)
- Gauri Tendulkar
- Siegfried Weller Institute for Trauma Research at the BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, Schnarrenbergstrasse 95, 72076 Tübingen, Germany
| | - Tao Chen
- Siegfried Weller Institute for Trauma Research at the BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, Schnarrenbergstrasse 95, 72076 Tübingen, Germany
| | - Sabrina Ehnert
- Siegfried Weller Institute for Trauma Research at the BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, Schnarrenbergstrasse 95, 72076 Tübingen, Germany
| | - Hans-Peter Kaps
- Siegfried Weller Institute for Trauma Research at the BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, Schnarrenbergstrasse 95, 72076 Tübingen, Germany
| | - Andreas K Nüssler
- Siegfried Weller Institute for Trauma Research at the BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, Schnarrenbergstrasse 95, 72076 Tübingen, Germany.
| |
Collapse
|
28
|
Dai J, Xing Y, Xiao L, Li J, Cao R, He Y, Fang H, Periasamy A, Oberhozler J, Jin L, Landers JP, Wang Y, Li X. Microfluidic Disc-on-a-Chip Device for Mouse Intervertebral Disc-Pitching a Next-Generation Research Platform To Study Disc Degeneration. ACS Biomater Sci Eng 2019; 5:2041-2051. [PMID: 31763444 DOI: 10.1021/acsbiomaterials.8b01522] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Low back pain is the most common cause of disability worldwide, and intervertebral disc degeneration is a major cause of low back pain. Unfortunately, discogenic low back pain is often treated with symptomatic relief interventions, as no disease-modifying medications are yet available. Both to-be-deciphered disc biology/pathology and inadequate in vitro research platform are major hurdles limiting drug discovery progress for disc degeneration. Here, we developed a microfluidic disc-on-a-chip device tailored for mouse disc organ as an in vitro research platform. We hypothesize that continuous nutrients empowered by a microfluidic device would improve biological performance of cultured mouse discs compared to those in static condition. This device permitted continuous media flow to mimic in vivo disc microenvironment. Intriguingly, mouse discs cultured on the microfluidic device exhibited much higher cell viability, better preserved structure integrity and anabolic-catabolic metabolism in both nucleus pulposus and annulus fibrosus, for up to 21 days compared to those in static culture. This first "disc-on-a-chip" device lays groundwork for future preclinical studies in a relative long-term organ culture given the chronic nature of intervertebral disc degeneration. In addition, this platform is readily transformable into a streamlined in vitro research platform to recapitulate physiological and pathophysiological microenvironment to accelerate disc research.
Collapse
Affiliation(s)
- Jun Dai
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Drive, Charlottesville, Virginia 22908, United States.,Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue Qiaokou District, Wuhan 430030, P.R. China
| | - Yuan Xing
- Department of Surgery, University of Virginia, 345 Cripell Drive, Charlottesville, Virginia 22908, United States
| | - Li Xiao
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Drive, Charlottesville, Virginia 22908, United States
| | - Jingyi Li
- ∥ Department of Chemistry, University of Virginia, 409 McCormick Road, Charlottesville, Virginia 22904, United States
| | - Ruofan Cao
- W.M. Keck Center for Cellular Imaging, University of Virginia, 90 Geldard Drive, Charlottesville, Virginia 22904, United States
| | - Yi He
- Department of Surgery, University of Virginia, 345 Cripell Drive, Charlottesville, Virginia 22908, United States
| | - Huang Fang
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue Qiaokou District, Wuhan 430030, P.R. China
| | - Ammasi Periasamy
- W.M. Keck Center for Cellular Imaging, University of Virginia, 90 Geldard Drive, Charlottesville, Virginia 22904, United States
| | - Jose Oberhozler
- Department of Surgery, University of Virginia, 345 Cripell Drive, Charlottesville, Virginia 22908, United States
| | - Li Jin
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Drive, Charlottesville, Virginia 22908, United States
| | - James P Landers
- ∥ Department of Chemistry, University of Virginia, 409 McCormick Road, Charlottesville, Virginia 22904, United States.,Department of Mechanical and Aerospace Engineering, University of Virginia, 122 Engineer's Way, Charlottesville, Virginia 22904, United States.,Department of Pathology, University of Virginia, 415 Lane Road, Charlottesville, Virginia 22908, United States
| | - Yong Wang
- Department of Surgery, University of Virginia, 345 Cripell Drive, Charlottesville, Virginia 22908, United States
| | - Xudong Li
- Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Drive, Charlottesville, Virginia 22908, United States.,Department of Biomedical Engineering, University of Virginia, 415 Lane Road, Charlottesville, Virginia 22908, United States
| |
Collapse
|
29
|
Torre OM, Das R, Berenblum RE, Huang AH, Iatridis JC. Neonatal mouse intervertebral discs heal with restored function following herniation injury. FASEB J 2018; 32:4753-4762. [PMID: 29570392 PMCID: PMC6103171 DOI: 10.1096/fj.201701492r] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/12/2018] [Indexed: 12/13/2022]
Abstract
Adult intervertebral discs (IVDs) have poor endogenous healing capacity, because of their challenging microenvironment and complex mechanical demands, which can result in painful IVD herniation. There are no regenerative strategies available to improve IVD healing and restore its function. Neonatal mice are excellent models of mammalian regeneration, but there are no studies of the regenerative capacity of neonatal IVDs. In this study, we developed a neonatal model of improved IVD healing to inform repair strategies after herniation. In vivo puncture injuries were performed to simulate herniation with complete annulus fibrosus (AF) tears in caudal IVDs of neonatal (postnatal d 5) and adult (4-6 mo) Scleraxis green fluorescent protein ( ScxGFP) mice. Acute and long-term healing responses were assessed with histologic, radiologic, and biomechanical measurements. Neonates underwent accelerated IVD healing compared to adults with functional restoration and enhanced structural repair after herniation. A population of ScxGFP- cells identified in the neonatal repair site may be associated with this improved healing and warrants future investigation. In summary, function of neonatal IVDs was restored after herniation injury, whereas that of adult discs was not. This improved healing response is likely driven by multiple mechanisms that may include differences in mechanical loading and available repair cells during growth.-Torre, O. M., Das, R., Berenblum, R. E., Huang, A. H., Iatridis, J. C. Neonatal mouse intervertebral discs heal with restored function following herniation injury.
Collapse
Affiliation(s)
- Olivia M. Torre
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rohit Das
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ramy E. Berenblum
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alice H. Huang
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - James C. Iatridis
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
30
|
Fang H, Li X, Shen H, Sun B, Teng H, Li P. Osteogenic protein-1 attenuates apoptosis and enhances matrix synthesis of nucleus pulposus cells under high-magnitude compression though inhibiting the p38 MAPK pathway. Biosci Rep 2018; 38:BSR20180018. [PMID: 29440560 PMCID: PMC5857901 DOI: 10.1042/bsr20180018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/05/2018] [Accepted: 02/12/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Disc degeneration is correlated with mechanical load. Osteogenic protein-1 (OP-1) is potential to regenerate degenerative disc. OBJECTIVE To investigate whether OP-1 can protect against high magitude compression-induced nucleus pulposus (NP) cell apoptosis and NP matrix catabolism, and its potential mechanism. METHODS Porcine discs were cultured in a bioreactor and compressed at a relatively high-magnitude mechanical compression (1.3 MPa at a frequency of 1.0 Hz for 2 hours once per day) for 7 days. OP-1 was added along with the culture medium to investigate the protective effects of OP-1. NP cell apoptosis and matrix biosynthesis were evaluated. Additionally, activity of the p38 MAPK pathway is also analyzed. RESULTS Compared with the control group, high magnitude compression significantly promoted NP cell apoptosis and decreased NP matrix biosynthesis, reflected by the increase in the number of TUNEL-positive cells and caspase-3 activity, the up-regulated expression of Bax and caspase-3 mRNA and down-regulated expression of Bcl-2 mRNA, and the decreased alcian blue staining intensity and expression of matrix proteins (aggrecan and collagen II). However, OP-1 addition partly attenuated the effects of high magnitude compression on NP cell apoptosis and NP matrix biosynthesis. Further analysis showed that inhibition of the p38 MAPK pathway partly participated in this process. CONCLUSION OP-1 can attenuate high magnitude compression-induced NP cell apoptosis and promoted NP matrix biosynthesis, and inhibition of the p38 MAPK pathway may participate in this regulatory process. This study provides that OP-1 may be efficacy in retarding mechanical overloading-exacerbated disc degeneration.
Collapse
Affiliation(s)
- Haolin Fang
- Department of Emergency Trauma Surgery, Jining No.1 People's Hospital, Jining 272011, P. R. China, Jining, China
| | - Xianzhou Li
- Department of Spinal Surgery, Jining No.1 People's Hospital, Jining 272011, P. R. China, Jining, China
| | - Haiming Shen
- Department of Neurosurgery, Jining No.2 People's Hospital, Jining 272011, P. R. China, Jining, China
| | - Buwei Sun
- Department of Orthopaedics, Jining No.2 People's Hospital, Jining 272011, P. R. China, Jining, China
| | - Haijun Teng
- Department of Orthopaedics, No.89 Hospital of PLA, Weifang, China
| | - Pei Li
- Department of Orthopedic Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
31
|
Nakazawa KR, Walter BA, Laudier DM, Krishnamoorthy D, Mosley GE, Spiller KL, Iatridis JC. Accumulation and localization of macrophage phenotypes with human intervertebral disc degeneration. Spine J 2018; 18:343-356. [PMID: 29031872 PMCID: PMC5815908 DOI: 10.1016/j.spinee.2017.09.018] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 09/18/2017] [Accepted: 09/26/2017] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Chronic inflammation is an important component of intervertebral disc (IVD) degeneration, but there is limited knowledge about the identity and source of inflammatory cells involved with the degenerative processes. Macrophages can exhibit multiple phenotypes and are known inflammatory regulators in many tissues, but their phenotypes have not been characterized in IVD degeneration. PURPOSE We aimed to characterize accumulation and localization of macrophages in IVD degeneration. STUDY DESIGN/SETTING This is an exploratory study to characterize macrophage phenotypes in human cadaver IVDs and the effects of injury and degeneration using multiple immunohistochemistry methods. OUTCOME MEASURES Percent positivity of immunohistochemical markers specific for CCR7, CD163, and CD206, and qualitative assessments of dual immunofluorescence and immunostaining localization were the outcome measures. METHODS Macrophages were identified in human cadaveric IVDs with immunohistochemistry using cell surface markers CCR7, CD163, and CD206, which are associated with proinflammatory M1, remodeling M2c, and anti-inflammatory M2a phenotypes, respectively. Variations in the accumulation and localization of macrophage markers with degenerative grade across subjects and within donors are described. RESULTS Cells expressing all three macrophage markers were found in all degenerative IVDs, but not in the healthiest IVDs. Cells expressing CCR7 and CD163, but not CD206, significantly increased with degenerative grade. Many cells also co-expressed multiple macrophage markers. Across all degenerative grades, CCR7+ and CD163+ were significantly more present in unhealthy nucleus pulposus (NP), annulus fibrosus (AF), and end plate (EP) regions exhibiting structural irregularities and defects. Positively stained cells in the NP and AF closely resembled resident IVD cells, suggesting that IVD cells can express macrophage cell surface markers. In the EP, there were increasing trends of positively stained cells with atypical morphology and distribution, suggesting a source for exogenous macrophage infiltration into the IVD. CONCLUSIONS Chronic inflammatory conditions of IVD degeneration appear to involve macrophages or macrophage-like cells, as expression of multiple macrophage markers increased with degeneration, especially around unhealthy regions with defects and the EP. Knowledge of macrophage phenotypes and their localization better elucidates the complex injury and repair processes in IVDs and may eventually lead to novel treatments.
Collapse
Affiliation(s)
- Kenneth R. Nakazawa
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Benjamin A. Walter
- Spine Research Institute, Department of Biomedical Engineering, The Ohio State University, Columbus, OH
| | - Damien M. Laudier
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Divya Krishnamoorthy
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Grace E. Mosley
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kara L. Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA
| | - James C. Iatridis
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
32
|
Xiao L, Hong K, Roberson C, Ding M, Fernandez A, Shen F, Jin L, Sonkusare S, Li X. Hydroxylated Fullerene: A Stellar Nanomedicine to Treat Lumbar Radiculopathy via Antagonizing TNF- α-Induced Ion Channel Activation, Calcium Signaling, and Neuropeptide Production. ACS Biomater Sci Eng 2017; 4:266-277. [PMID: 30038959 DOI: 10.1021/acsbiomaterials.7b00735] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Current nonsurgical treatments of discogenic lumbar radiculopathy are neither effective nor safe. Our prior studies have suggested that hydroxylated fullerene (fullerol) nanomaterial could attenuate proinflammatory cytokine tumor necrosis factor alpha (TNF-α)-induced neuroinflammation and oxidative stress in mouse dorsal root ganglia (DRG) and primary neurons. Here, we aim to investigate the analgesic effect of fullerol in a clinically relevant lumbar radiculopathy mouse model and to understand its underlying molecular mechanism in mouse DRGs and neurons. Surprisingly, single and local application of fullerol solution (1 μM, 10 μL) was sufficient to alleviate ipsilateral paw pain sensation in mice up to 2 weeks postsurgery. In addition, microCT data suggested fullerol potentially promoted disc height recovery following injury-induced disc herniation. Alcian blue/picrosirius red staining also suggested that fullerol promoted regeneration of extracellular matrix proteins visualized by the presence of abundant newly formed collagen and proteoglycan in herniated discs. For in vitro DRG culture, fullerol attenuated TNF-α-elicited expression of transient receptor potential cation channel subfamily V member 1 (TRPV-1) and neuropeptides release (substance P and calcitonin gene-related peptide). In addition, fullerol suppressed TNF-α-stimulated increase in intracellular Ca2+ concentrations in primary neurons. Moreover, Western blot analysis in DRG revealed that fullerol's beneficial effects against TNF-α might be mediated through protein kinase B (AKT) and extracellular protein-regulated kinase (ERK) pathways. These TNF-α antagonizing and analgesic effects indicated therapeutic potential of fullerol in treating lumbar radiculopathy, providing solid preclinical evidence toward further translational studies.
Collapse
Affiliation(s)
- Li Xiao
- Department of Orthopaedic Surgery, Charlottesville, Virginia 22908, United States
| | - Kwangseok Hong
- Robert M. Berne Cardiovascular Research Center, Charlottesville, Virginia 22908, United States
| | - Charles Roberson
- Department of Orthopaedic Surgery, Charlottesville, Virginia 22908, United States
| | - Mengmeng Ding
- Department of Orthopaedic Surgery, Charlottesville, Virginia 22908, United States
| | - Andrew Fernandez
- Department of Orthopaedic Surgery, Charlottesville, Virginia 22908, United States
| | - Francis Shen
- Department of Orthopaedic Surgery, Charlottesville, Virginia 22908, United States
| | - Li Jin
- Department of Orthopaedic Surgery, Charlottesville, Virginia 22908, United States
| | - Swapnil Sonkusare
- Robert M. Berne Cardiovascular Research Center, Charlottesville, Virginia 22908, United States.,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Xudong Li
- Department of Orthopaedic Surgery, Charlottesville, Virginia 22908, United States.,Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, United States
| |
Collapse
|
33
|
Chitosan oligosaccharides protect nucleus pulposus cells from hydrogen peroxide-induced apoptosis in a rat experimental model. Biomed Pharmacother 2017; 93:807-815. [DOI: 10.1016/j.biopha.2017.06.101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 06/12/2017] [Accepted: 06/29/2017] [Indexed: 01/06/2023] Open
|
34
|
Xiao L, Ding M, Fernandez A, Zhao P, Jin L, Li X. Curcumin alleviates lumbar radiculopathy by reducing neuroinflammation, oxidative stress and nociceptive factors. Eur Cell Mater 2017; 33:279-293. [PMID: 28485773 PMCID: PMC5521990 DOI: 10.22203/ecm.v033a21] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Current non-surgical treatments for lumbar radiculopathy [e.g. epidural steroids and Tumour necrosis factor-α (TNF-α) antagonists] are neither effective nor safe. As a non-toxic natural product, curcumin possesses an exceptional anti-inflammatory profile. We hypothesised that curcumin alleviates lumbar radiculopathy by attenuating neuroinflammation, oxidative stress and nociceptive factors. In a dorsal root ganglion (DRG) culture, curcumin effectively inhibited TNF-α-induced neuroinflammation, in a dose-dependent manner, as shown by mRNA and protein expression of IL-6 and COX-2. Such effects might be mediated via protein kinase B (AKT) and extracellular signal regulated kinase (ERK) pathways. Also, a similar effect in combating TNF-α-induced neuroinflammation was observed in isolated primary neurons. In addition, curcumin protected neurons from TNF-α-triggered excessive reactive oxygen species (ROS) production and cellular apoptosis and, accordingly, promoted mRNA expression of the anti-oxidative enzymes haem oxygenase-1, catalase and superoxide dismutase-2. Intriguingly, electronic von Frey test suggested that intraperitoneal injection of curcumin significantly abolished ipsilateral hyperalgesia secondary to disc herniation in mice, for up to 2 weeks post-surgery. Such in vivo pain alleviation could be attributed to the suppression, observed in DRG explant culture, of TNF-α-elicited neuropeptides, such as substance P and calcitonin gene-related peptide. Surprisingly, micro-computed tomography (μCT) data suggested that curcumin treatment could promote disc height recovery following disc herniation. Alcian blue/picrosirius red staining confirmed that systemic curcumin administration promoted regeneration of extracellular matrix proteins, visualised by presence of abundant newly-formed collagen and proteoglycan content in herniated disc. Our study provided pre-clinical evidence for expediting this natural, non-toxic pleiotropic agent to become a new and safe clinical treatment of radiculopathy.
Collapse
Affiliation(s)
- L. Xiao
- Department of Orthopaedic Surgery, University of Virginia, Cobb Hall, 135 Hospital Dr. Charlottesville, VA 22908, USA
| | - M. Ding
- Department of Orthopaedic Surgery, University of Virginia, Cobb Hall, 135 Hospital Dr. Charlottesville, VA 22908, USA,Department of Anaesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - A. Fernandez
- Department of Orthopaedic Surgery, University of Virginia, Cobb Hall, 135 Hospital Dr. Charlottesville, VA 22908, USA
| | - P. Zhao
- Department of Anaesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - L. Jin
- Department of Orthopaedic Surgery, University of Virginia, Cobb Hall, 135 Hospital Dr. Charlottesville, VA 22908, USA
| | - X. Li
- Department of Orthopaedic Surgery, University of Virginia, Cobb Hall, 135 Hospital Dr. Charlottesville, VA 22908, USA,Address for correspondence: Dr Xudong Li, MD, PhD, Rm B051, Cobb Hall, Department of Orthopaedic Surgery, University of Virginia, 135 Hospital Dr. Charlottesville, VA 22908, USA, Telephone number: +1 4349824135, Fax number: +1 4349241691,
| |
Collapse
|
35
|
Long RG, Rotman SG, Hom WW, Assael DJ, Illien-Jünger S, Grijpma DW, Iatridis JC. In vitro and biomechanical screening of polyethylene glycol and poly(trimethylene carbonate) block copolymers for annulus fibrosus repair. J Tissue Eng Regen Med 2017; 12:e727-e736. [PMID: 27860368 DOI: 10.1002/term.2356] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/08/2016] [Accepted: 11/09/2016] [Indexed: 12/23/2022]
Abstract
Herniated intervertebral discs (IVDs) are a common cause of back and neck pain. There is an unmet clinical need to seal annulus fibrosus (AF) defects, as discectomy surgeries address acute pain but are complicated by reherniation and recurrent pain. Copolymers of polyethylene glycol with trimethylene carbonate (TMC) and hexamethylene diisocyanate (HDI) end-groups were formulated as AF sealants as the HDI form covalent bonds with native AF tissue. TMC adhesives were evaluated and optimized using the design criteria: stable size, strong adherence to AF tissue, high cytocompatibility, restoration of IVD biomechanics to intact levels following in situ repair, and low extrusion risk. TMC adhesives had high adhesion strength as assessed with a pushout test (150 kPa), and low degradation rates over 3 weeks in vitro. Both TMC adhesives had shear moduli (220 and 490 kPa) similar to, but somewhat higher than, AF tissue. The adhesive with three TMC moieties per branch (TMC3) was selected for additional in situ testing because it best matched AF shear properties. TMC3 restored torsional stiffness, torsional hysteresis area and axial range of motion to intact states. However, in a failure test of compressive deformation under fixed 5 ° flexion, some herniation risk was observed with failure strength of 5.9 MPa compared with 13.5 MPa for intact samples; TMC3 herniated under cyclic organ culture testing. These TMC adhesives performed well during in vitro and in situ testing, but additional optimization to enhance failure strength is required to further this material to advanced screening tests, such as long-term degradation. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Rose G Long
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - Stijn G Rotman
- Department of Biomaterials Science and Technology, University of Twente, Enschede, the Netherlands
| | - Warren W Hom
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dylan J Assael
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Svenja Illien-Jünger
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dirk W Grijpma
- Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland.,Department of Biomaterials Science and Technology, University of Twente, Enschede, the Netherlands.,Department of Biomedical Engineering, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - James C Iatridis
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| |
Collapse
|
36
|
Walter B, Purmessur D, Moon A, Occhiogrosso J, Laudier D, Hecht A, Iatridis J. Reduced tissue osmolarity increases TRPV4 expression and pro-inflammatory cytokines in intervertebral disc cells. Eur Cell Mater 2016; 32:123-36. [PMID: 27434269 PMCID: PMC5072776 DOI: 10.22203/ecm.v032a08] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mechanical behaviour and cellular metabolism of intervertebral discs (IVDs) and articular cartilage are strongly influenced by their proteoglycan content and associated osmotic properties. This osmotic environment is a biophysical signal that changes with disease and may contribute to the elevated matrix breakdown and altered biologic response to loading observed in IVD degeneration and osteoarthritis. This study tested the hypothesis that changes in osmo-sensation by the transient receptor potential vallinoid-4 (TRPV4) ion channel occur with disease and contribute to the inflammatory environment found during degeneration. Immunohistochemistry on bovine IVDs from an inflammatory organ culture model were used to investigate if TRPV4 is expressed in the IVD and how expression changes with degeneration. Western blot, live-cell calcium imaging, and qRT-PCR were used to investigate whether osmolarity changes or tumour necrosis factor α (TNFα) regulate TRPV4 expression, and how altered TRPV4 expression influences calcium signalling and pro-inflammatory cytokine expression. TRPV4 expression correlated with TNFα expression, and was increased when cultured in reduced medium osmolarity and unaltered with TNFα-stimulation. Increased TRPV4 expression increased the calcium flux following TRPV4 activation and increased interleukin-1β (IL-1β) and IL-6 gene expression in IVD cells. TRPV4 expression was qualitatively elevated in regions of aggrecan depletion in degenerated human IVDs. Collectively, results suggest that reduced tissue osmolarity, likely following proteoglycan degradation, can increase TRPV4 signalling and enhance pro-inflammatory cytokine production, suggesting changes in TRPV4 mediated osmo-sensation may contribute to the progressive matrix breakdown in disease.
Collapse
Affiliation(s)
- B.A. Walter
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - D Purmessur
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - A. Moon
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J. Occhiogrosso
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D.M. Laudier
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - A.C. Hecht
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J.C. Iatridis
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA,Address for correspondence: James C. Iatridis Leni & Peter W. May Department of Orthopaedics, Box 1188, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA, Telephone Number: 1-212-241-1517, FAX Number: 1-212-876-3168 www.ecmjournal.org
| |
Collapse
|
37
|
The Natural Polyphenol Epigallocatechin Gallate Protects Intervertebral Disc Cells from Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:7031397. [PMID: 27119009 PMCID: PMC4826942 DOI: 10.1155/2016/7031397] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/30/2016] [Accepted: 02/16/2016] [Indexed: 12/19/2022]
Abstract
Oxidative stress-related phenotypic changes and a decline in the number of viable cells are crucial contributors to intervertebral disc degeneration. The polyphenol epigallocatechin 3-gallate (EGCG) can interfere with painful disc degeneration by reducing inflammation, catabolism, and pain. In this study, we hypothesized that EGCG furthermore protects against senescence and/or cell death, induced by oxidative stress. Sublethal and lethal oxidative stress were induced in primary human intervertebral disc cells with H2O2 (total n = 36). Under sublethal conditions, the effects of EGCG on p53-p21 activation, proliferative capacity, and accumulation of senescence-associated β-galactosidase were tested. Further, the effects of EGCG on mitochondria depolarization and cell viability were analyzed in lethal oxidative stress. The inhibitor LY249002 was applied to investigate the PI3K/Akt pathway. EGCG inhibited accumulation of senescence-associated β-galactosidase but did not affect the loss of proliferative capacity, suggesting that EGCG did not fully neutralize exogenous radicals. Furthermore, EGCG increased the survival of IVD cells in lethal oxidative stress via activation of prosurvival PI3K/Akt and protection of mitochondria. We demonstrated that EGCG not only inhibits inflammation but also can enhance the survival of disc cells in oxidative stress, which makes it a suitable candidate for the development of novel therapies targeting disc degeneration.
Collapse
|
38
|
Long RG, Bürki A, Zysset P, Eglin D, Grijpma DW, Blanquer SBG, Hecht AC, Iatridis JC. Mechanical restoration and failure analyses of a hydrogel and scaffold composite strategy for annulus fibrosus repair. Acta Biomater 2016; 30:116-125. [PMID: 26577987 DOI: 10.1016/j.actbio.2015.11.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 11/04/2015] [Accepted: 11/10/2015] [Indexed: 01/07/2023]
Abstract
Unrepaired defects in the annulus fibrosus of intervertebral disks are associated with degeneration and persistent back pain. A clinical need exists for a disk repair strategy that can seal annular defects, be easily delivered during surgical procedures, and restore biomechanics with low risk of herniation. Multiple annulus repair strategies were developed using poly(trimethylene carbonate) scaffolds optimized for cell delivery, polyurethane membranes designed to prevent herniation, and fibrin-genipin adhesive tuned to annulus fibrosus shear properties. This three-part study evaluated repair strategies for biomechanical restoration, herniation risk and failure mode in torsion, bending and compression at physiological and hyper-physiological loads using a bovine injury model. Fibrin-genipin hydrogel restored some torsional stiffness, bending ROM and disk height loss, with negligible herniation risk and failure was observed histologically at the fibrin-genipin mid-substance following rigorous loading. Scaffold-based repairs partially restored biomechanics, but had high herniation risk even when stabilized with sutured membranes and failure was observed histologically at the interface between scaffold and fibrin-genipin adhesive. Fibrin-genipin was the simplest annulus fibrosus repair solution evaluated that involved an easily deliverable adhesive that filled irregularly-shaped annular defects and partially restored disk biomechanics with low herniation risk, suggesting further evaluation for disk repair may be warranted. STATEMENT OF SIGNIFICANCE Lower back pain is the leading cause of global disability and commonly caused by defects and failure of intervertebral disk tissues resulting in herniation and compression of adjacent nerves. Annulus fibrosus repair materials and techniques have not been successful due to the challenging mechanical and chemical microenvironment and the needs to restore biomechanical behaviors and promote healing with negligible herniation risk while being delivered during surgical procedures. This work addressed this challenging biomaterial and clinical problem using novel materials including an adhesive hydrogel, a scaffold capable of cell delivery, and a membrane to prevent herniation. Composite repair strategies were evaluated and optimized in quantitative three-part study that rigorously evaluated disk repair and provided a framework for evaluating alternate repair techniques.
Collapse
Affiliation(s)
- Rose G Long
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - Alexander Bürki
- Institute for Surgical Technology & Biomechanics, University of Bern, Bern, Switzerland
| | - Philippe Zysset
- Institute for Surgical Technology & Biomechanics, University of Bern, Bern, Switzerland
| | - David Eglin
- AO Research Institute Davos, Davos, Switzerland; Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - Dirk W Grijpma
- Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland; University of Twente, Department of Biomaterials Science and Technology, PO Box 217, 7500 AE Enschede, The Netherlands; University of Groningen, University Medical Centre Groningen, Department of Biomedical Engineering, PO Box 196, 9700 AD Groningen, The Netherlands
| | - Sebastien B G Blanquer
- Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland; University of Twente, Department of Biomaterials Science and Technology, PO Box 217, 7500 AE Enschede, The Netherlands
| | - Andrew C Hecht
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James C Iatridis
- Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland.
| |
Collapse
|