1
|
Wasman Smail S, Ziyad Abdulqadir S, Omar Khudhur Z, Elia Ishaq S, Faqiyazdin Ahmed A, Ghayour MB, Abdolmaleki A. IL-33 promotes sciatic nerve regeneration in mice by modulating macrophage polarization. Int Immunopharmacol 2023; 123:110711. [PMID: 37531832 DOI: 10.1016/j.intimp.2023.110711] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/11/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
Despite the innate regenerative capacity of peripheral nerves, regeneration after a severe injury is insufficient, and sensorimotor recovery is incomplete. As a result, finding alternative methods for improving regeneration and sensorimotor recovery is essential. In this regard, we investigated the effect of IL-33 treatment as a chemokine with neuroprotective properties. IL-33 can facilitate tissue healing by potentiating the type 2 immune response and polarizing macrophages toward the pro-healing M2 phenotype. However, its effects on nerve regeneration remain unclear. Therefore, this research aimed to evaluate the neuroprotective effects of IL-33 on sciatic nerve injury in male C57BL/6 mice. After crushing the left sciatic nerve, the animals were given 10, 25, or 50 µg/kg IL-33 intraperitoneally for seven days. The sensorimotor recovery was then assessed eight weeks after surgery. In addition, immunohistochemistry, ELISA, and real-time PCR were used to assess macrophage polarization, cytokine secretion, and neurotrophic factor expression in the injured nerves. IL-33 at 50 and 25 µg/kg doses could significantly accelerate nerve regeneration and improve sensorimotor recovery when compared to 10 µg/kg IL-33 and control groups. Furthermore, at 50 and 25 µg/kg doses, IL-33 polarized macrophages toward an M2 phenotype and reduced proinflammatory cytokines at the injury site. It also increased the mRNA expression of NGF, VEGF, and BDNF. These findings suggest that a seven-day IL-33 treatment had neuroprotective effects in a mouse sciatic nerve crush model, most likely by inducing macrophage polarization toward M2 and regulating inflammatory microenvironments.
Collapse
Affiliation(s)
- Shukur Wasman Smail
- Department of Medical Microbiology, College of Science, Cihan University-Erbil, Kurdistan Region, Iraq; Department of Biology, College of Science, Salahaddin University-Erbil, Kurdistan Region, Iraq
| | - Shang Ziyad Abdulqadir
- Department of Biology, College of Science, Salahaddin University-Erbil, Kurdistan Region, Iraq
| | - Zhikal Omar Khudhur
- Department of Biology Education, Faculty of Education, Tishk International University - Erbil, Kurdistan Region, Iraq.
| | - Sonia Elia Ishaq
- Department of Biology, College of Science, Salahaddin University-Erbil, Kurdistan Region, Iraq
| | | | - Mohammad B Ghayour
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Arash Abdolmaleki
- Department of Biophysics, Faculty of Advanced Technologies, University of Mohaghegh Ardabili, Namin, Iran.
| |
Collapse
|
2
|
Aman M, Mayrhofer-Schmid M, Schwarz D, Bendszus M, Daeschler SC, Klemm T, Kneser U, Harhaus L, Boecker AH. Avoiding scar tissue formation of peripheral nerves with the help of an acellular collagen matrix. PLoS One 2023; 18:e0289677. [PMID: 37540691 PMCID: PMC10403074 DOI: 10.1371/journal.pone.0289677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/24/2023] [Indexed: 08/06/2023] Open
Abstract
INTRODUCTION Extensive scar tissue formation after peripheral nerve injury or surgery is a common problem. To avoid perineural scarring, implanting a mechanical barrier protecting the nerve from inflammation processes in the perineural environment has shown promising results for functional recovery. This study investigates the potential of an acellular collagen-elastin matrix wrapped around a peripheral nerve after induction of scar tissue formation. MATERIALS AND METHODS In the present study, 30 Lewis rats were separated into three groups and sciatic nerve scarring was induced with 2.5% glutaraldehyde (GA-CM) or 2.5% glutaraldehyde with a supplemental FDA-approved acellular collagen-elastin matrix application (GA+CM). Additionally, a sham group was included for control. Nerve regeneration was assessed by functional analysis using the Visual Statisc Sciatic Index (SSI) and MR neurography during the 12-week regeneration period. Histological and histomorphometry analysis were performed to evaluate the degree of postoperative scar tissue formation. RESULTS Histological analysis showed an extensive scar tissue formation for GA-CM. Connective tissue ratio was significantly (p < 0.009) reduced for GA+CM (1.347 ± 0.017) compared to GA-CM (1.518 ± 0.057). Similarly, compared to GA+CM, MR-Neurography revealed extensive scar tissue formation for GA-CM with a direct connection between nerve and paraneural environment. Distal to the injury site, quantitative analysis presented significantly higher axon density (p = 0.0145), thicker axon diameter (p = 0.0002) and thicker myelinated fiber thickness (p = 0.0008) for GA+CM compared to GA-CM. Evaluation of functional recovery revealed a significantly faster regeneration for GA+CM. CONCLUSION The supplemental application of an acellular collagen-elastin matrix showed beneficial effects in histological, radiological, and functional analysis. Therefore, applying a collagen-elastin matrix around the nerve after peripheral nerve injury or surgery may have beneficial effects on preventing scar tissue formation in the long run. This represents a feasible approach to avoid scar tissue formation in peripheral nerve surgery.
Collapse
Affiliation(s)
- Martin Aman
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Hand- and Plastic Surgery, University of Heidelberg, Heidelberg, Germany
| | - Maximilian Mayrhofer-Schmid
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Hand- and Plastic Surgery, University of Heidelberg, Heidelberg, Germany
| | - Daniel Schwarz
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Simeon C Daeschler
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Hand- and Plastic Surgery, University of Heidelberg, Heidelberg, Germany
| | - Tess Klemm
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Hand- and Plastic Surgery, University of Heidelberg, Heidelberg, Germany
| | - Ulrich Kneser
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Hand- and Plastic Surgery, University of Heidelberg, Heidelberg, Germany
| | - Leila Harhaus
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Hand- and Plastic Surgery, University of Heidelberg, Heidelberg, Germany
| | - Arne H Boecker
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Department of Hand- and Plastic Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
3
|
Al-Arbeed TA, Renno WM, Al-Hassan JM. Neuroregeneration of injured peripheral nerve by fraction B of catfish epidermal secretions through the reversal of the apoptotic pathway and DNA damage. Front Pharmacol 2023; 14:1085314. [PMID: 36726586 PMCID: PMC9885176 DOI: 10.3389/fphar.2023.1085314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/03/2023] [Indexed: 01/17/2023] Open
Abstract
Introduction: Crush injuries occur from acute traumatic nerve compression resulting in different degrees of neural damage leading to permanent functional deficits. Recently, we have shown that administration of Fraction B (FB) derived from catfish epidermal secretions accelerates healing of damaged nerve in a sciatic nerve crush injury, as it ameliorates the neurobehavioral deficits and enhances axonal regeneration, as well as protects spinal neurons and increases astrocytic activity and decreasing GAP-43 expression. The present study aimed to investigate the role of FB treatment on the apoptotic pathway in the neuroregeneration of the sciatic nerve crush injury. Methods: Male Wistar rats were randomly assigned into five groups: (I) SHAM, (II) CRUSH, (III) CRUSH + (1.5 mg/kg) FB, (IV) CRUSH + (3 mg/kg) FB, and (V) CRUSH + (4.5 mg/kg) FB. Rats underwent sciatic nerve crush surgery, followed by treatment with FB administered intraperitoneally (IP) daily for two weeks and then sacrificed at the end of the fourth week. Results: FB improved the recovery of neurobehavioral functions with a concomitant increase in axonal regeneration and neuroprotective effects on spinal cord neurons following crush injury. Further, FB enhanced Schwann cells (SCs) proliferation with a significant increase in myelin basic protein expression. FB-treated animals demonstrated higher numbers of neurons in the spinal cord, possibly through ameliorating oxidative DNA damage and alleviating the mitochondrial-dependent apoptotic pathway by inhibiting the release of cytochrome c and the activation of caspase-3 in the spinal cord neurons. Conclusion: FB alleviates the neurodegenerative changes in the lumbar spinal cord neurons and recovers the decrease in the neuronal count through its anti-apoptotic and DNA antioxidative properties.
Collapse
Affiliation(s)
- Taiba A. Al-Arbeed
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Waleed M. Renno
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait,*Correspondence: Waleed M. Renno,
| | - Jassim M. Al-Hassan
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
4
|
Çalışkan SG, Bilgin MD. Nonlinear surface EMG analysis to detect the neuroprotective effect of citicoline in rat sciatic nerve crush injury. Med Biol Eng Comput 2022; 60:2865-2875. [DOI: 10.1007/s11517-022-02639-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 07/28/2022] [Indexed: 12/01/2022]
|
5
|
Bayır UÖ, Aksu R, Öz Gergin Ö, Onder GO, Sencar L, Günay E, Yay AH, Karaman İ, Bicer C, Polat S. The effect of pulsed radiofrequency application on nerve healing after sciatic nerve anastomosis in rats. Ultrastruct Pathol 2022; 46:313-322. [PMID: 35866415 DOI: 10.1080/01913123.2022.2066237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The Effect of Pulsed Radiofrequency Application on Nerve Healing After Sciatic Nerve Anastomosis in Rats. In this study, we aimed to evaluate the histomorphological and functional effect of Pulsed Radiofrequency (PRF) application on regeneration after experimental nerve damage in rats. Forty Sprague-Dawley male rats were used in the study. Sciatic nerve incision was applied to all rats and then anastomosis was performed. Twenty rats were separated as the control group, and the remaining 20 rats underwent PRF every day at 42oC, for 120 seconds. The groups were divided into two further subgroups to be sacrificed on the 15th and 30th days. Tissue samples were obtained from all groups at 24 hours and 72 hours after the injury. Sections of sciatic nerve samples were stained with hematoxylin-eosin for light microscopic investigation and prepared for evaluation of ultrastructural changes with transmission electron microscopy. In the evaluation of axon numbers and diameters were seen that the 30th-day RF group had an increase compared to the control group. In the electron microscopic examination, it was observed that myelinated and unmyelinated nerve fiber sheaths had borders that are more regular in the RF group, the nucleus structures of schwann cells were better preserved, mitochondrial damage was less, and the extensions of fibroblast and collagen fibers were smoother than the control group. The findings suggested that PRF application has a positive contribution histologically on nerve healing in the early period after full-layer incision nerve injury anastomosis surgery.
Collapse
Affiliation(s)
- Uğur Ö Bayır
- Department of Anesthesiology and Reanimation, Yozgat State Hospital, Yozgat, Turkey
| | - Recep Aksu
- Department of Anesthesiology and Reanimation, Medical Faculty, Erciyes University, Kayseri, Turkey
| | - Özlem Öz Gergin
- Department of Anesthesiology and Reanimation, Medical Faculty, Erciyes University, Kayseri, Turkey
| | - Gozde Ozge Onder
- Department of Histology and Embryology, Medical Faculty, Erciyes University, Kayseri, Turkey
| | - Leman Sencar
- Department of Histology and Embryology, Cukurova University, Medical Faculty, Adana, Turkey
| | - Eray Günay
- Department of Orthopaedic Surgery and Traumatology, Medical Faculty, Erciyes University, Kayseri, Turkey
| | - Arzu H Yay
- Department of Histology and Embryology, Medical Faculty, Erciyes University, Kayseri, Turkey.,Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| | - İbrahim Karaman
- Department of Orthopaedic Surgery and Traumatology, Medical Faculty, Erciyes University, Kayseri, Turkey
| | - Cihangir Bicer
- Department of Anesthesiology and Reanimation, Medical Faculty, Erciyes University, Kayseri, Turkey
| | - Sait Polat
- Department of Histology and Embryology, Cukurova University, Medical Faculty, Adana, Turkey
| |
Collapse
|
6
|
Alvites RD, Branquinho MV, Sousa AC, Lopes B, Sousa P, Prada J, Pires I, Ronchi G, Raimondo S, Luís AL, Geuna S, Varejão ASP, Maurício AC. Effects of Olfactory Mucosa Stem/Stromal Cell and Olfactory Ensheating Cells Secretome on Peripheral Nerve Regeneration. Biomolecules 2022; 12:biom12060818. [PMID: 35740943 PMCID: PMC9220795 DOI: 10.3390/biom12060818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 12/15/2022] Open
Abstract
Cell secretome has been explored as a cell-free technique with high scientific and medical interest for Regenerative Medicine. In this work, the secretome produced and collected from Olfactory Mucosa Mesenchymal Stem Cells and Olfactory Ensheating Cells was analyzed and therapeutically applied to promote peripheral nerve regeneration. The analysis of the conditioned medium revealed the production and secretion of several factors with immunomodulatory functions, capable of intervening beneficially in the phases of nerve regeneration. Subsequently, the conditioned medium was applied to sciatic nerves of rats after neurotmesis, using Reaxon® as tube-guides. Over 20 weeks, the animals were subjected to periodic functional assessments, and after this period, the sciatic nerves and cranial tibial muscles were evaluated stereologically and histomorphometrically, respectively. The results obtained allowed to confirm the beneficial effects resulting from the application of this therapeutic combination. The administration of conditioned medium from Olfactory Mucosal Mesenchymal Stem Cells led to the best results in motor performance, sensory recovery, and gait patterns. Stereological and histomorphometric evaluation also revealed the ability of this therapeutic combination to promote nervous and muscular histologic reorganization during the regenerative process. The therapeutic combination discussed in this work shows promising results and should be further explored to clarify irregularities found in the outcomes and to allow establishing the use of cell secretome as a new therapeutic field applied in the treatment of peripheral nerves after injury.
Collapse
Affiliation(s)
- Rui D. Alvites
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
| | - Mariana V. Branquinho
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
| | - Ana C. Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
| | - Bruna Lopes
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
| | - Patrícia Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
| | - Justina Prada
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
- Centro de Ciência Animal e Veterinária (CECAV), Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
| | - Isabel Pires
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
- Centro de Ciência Animal e Veterinária (CECAV), Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
| | - Giulia Ronchi
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (G.R.); (S.R.); (S.G.)
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (G.R.); (S.R.); (S.G.)
| | - Ana L. Luís
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (G.R.); (S.R.); (S.G.)
| | - Artur Severo P. Varejão
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
- Centro de Ciência Animal e Veterinária (CECAV), Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
| | - Ana Colette Maurício
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (A.L.L.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (A.S.P.V.)
- Correspondence: ; Tel.: +351-91-9071286 or +351-22-0428000
| |
Collapse
|
7
|
Weiss JB, Phillips CJ, Malin EW, Gorantla VS, Harding JW, Salgar SK. Stem cell, Granulocyte-Colony Stimulating Factor and/or Dihexa to promote limb function recovery in a rat sciatic nerve damage-repair model: Experimental animal studies. Ann Med Surg (Lond) 2021; 71:102917. [PMID: 34703584 PMCID: PMC8524106 DOI: 10.1016/j.amsu.2021.102917] [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: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 11/04/2022] Open
Abstract
Background Optimizing nerve regeneration and re-innervation of target muscle/s is the key for improved functional recovery following peripheral nerve damage. We investigated whether administration of mesenchymal stem cell (MSC), Granulocyte-Colony Stimulating Factor (G-CSF) and/or Dihexa can improve recovery of limb function following peripheral nerve damage in rat sciatic nerve transection-repair model. Materials and methods There were 10 experimental groups (n = 6–8 rats/group). Bone marrow derived syngeneic MSCs (2 × 106; passage≤6), G-CSF (200–400 μg/kg b.wt.), Dihexa (2–4 mg/kg b.wt.) and/or Vehicle were administered to male Lewis rats locally via hydrogel at the site of nerve repair, systemically (i.v./i.p), and/or to gastrocnemius muscle. The limb sensory and motor functions were assessed at 1–2 week intervals post nerve repair until the study endpoint (16 weeks). Results The sensory function in all nerve boundaries (peroneal, tibial, sural) returned to nearly normal by 8 weeks (Grade 2.7 on a scale of Grade 0–3 [0 = No function; 3 = Normal function]) in all groups combined. The peroneal nerve function recovered quickly with return of function at one week (∼2.0) while sural nerve function recovered rather slowly at four weeks (∼1.0). Motor function at 8–16 weeks post-nerve repair as determined by walking foot print grades significantly (P < 0.05) improved with MSC + G-CSF or MSC + Dihexa administrations into gastrocnemius muscle and mitigated foot flexion contractures. Conclusions These findings demonstrate MSC, G-CSF and Dihexa are promising candidates for adjunct therapies to promote limb functional recovery after surgical nerve repair, and have implications in peripheral nerve injury and limb transplantation. IACUC No.215064. G-CSF in combination with MSCs improved limb function recovery in sciatic nerve transection- repair model. Dihexa in combination with MSC improved limb function recovery in sciatic nerve transection- repair model. Foot flexion contractures were reduced with G-CSF & MSC or Dihexa & MSC administration into target muscle gastrocnemius. MSC, G-CSF or Dihexa combination therapy is attractive, feasible & promising in peripheral nerve injury repair and have implications in limb transplantation. The findings warrant further investigation to understand the cellular/molecular mechanisms.
Collapse
Affiliation(s)
- Jessica B Weiss
- Department of Surgery, Madigan Army Medical Center, Tacoma, Fort Lewis, Washington, USA
| | - Cody J Phillips
- Department of Surgery, Madigan Army Medical Center, Tacoma, Fort Lewis, Washington, USA
| | - Edward W Malin
- Department of Surgery, Madigan Army Medical Center, Tacoma, Fort Lewis, Washington, USA
| | - Vijay S Gorantla
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Joseph W Harding
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA, USA
| | - Shashikumar K Salgar
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, Fort Lewis, Washington, USA
| |
Collapse
|
8
|
Renno WM, Afzal M, Paul B, Nair D, Kumar J, Al-Hassan JM. Catfish Epidermal Preparation Accelerates Healing of Damaged Nerve in a Sciatic Nerve Crush Injury Rat Model. Front Pharmacol 2021; 12:632028. [PMID: 33986668 PMCID: PMC8112254 DOI: 10.3389/fphar.2021.632028] [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: 11/21/2020] [Accepted: 01/18/2021] [Indexed: 11/13/2022] Open
Abstract
Preliminary investigations showed that preparations from Arabian Gulf catfish (Arius bilineatus, Val) epidermal gel secretion (PCEGS) exhibit potent anti-inflammatory and healing properties as shown in our previous clinical trials for the healing of non-healing diabetic foot ulcers, chronic back pain, and some other neurological disorders. Here, we report for the first time a unique preparation containing only proteins and lipids (soluble protein fraction B, SPF-FB), derived from the PCEGS accelerated the healing and recovery of sensory-motor functions of experimental sciatic nerve crush injury in rats with its unique neuroprotective and neuroregenerative properties on the spinal neurons and peripheral nerve fibers. Male rats were randomly assigned to five groups: (I) NAÏVE, (II) SHAM, (III) CRUSH treated with saline, (IV) CRUSH + SPF-FB treated with 3 mg/kg intraperitoneally (IP) and (V) CRUSH + SPF-FB treated with 6 mg/kg subcutaneously (SC) groups. The crush groups III, IV and V underwent sciatic nerve crush injury, followed by treatment daily for 14 days with saline, SPF-FB IP and SPF-FB SC. All animals were tested for the neurobehavioral parameters throughout the 6 weeks of the study. Sciatic nerve and spinal cord tissues were processed for light and electron histological examinations, stereological analysis, immunohistochemical and biochemical examinations at Week 4 and Week 6 post-injury. Administration of SPF-FB IP or SC significantly enhanced the neurobehavioral sensory and motor performance and histomorphological neuroregeneration of the sciatic nerve-injured rats. The stereological evaluation of the axon area, average axon perimeters, and myelin thickness revealed significant histomorphological evidence of neuroregeneration in the FB-treated sciatic nerve crush injured groups compared to controls at 4 and 6 weeks. SPF-FB treatment significantly prevented the increased in NeuN-immunoreactive neurons, increased GFAP immunoreactive astrocytes, and decreased GAP-43. We conclude that SPF-FB treatment lessens neurobehavioral deficits, enhances axonal regeneration following nerve injury. We conclude that SPF-FB treatment lessens neurobehavioral deficits and enhances axonal regeneration following nerve injury, as well as protects spinal neurons and enhances subcellular recovery by increasing astrocytic activity and decreasing GAP-43 expression.
Collapse
Affiliation(s)
- Waleed M Renno
- Department of Anatomy, Faculty of Medicine, Kuwait University, Safat, Kuwait
| | - Mohammad Afzal
- Biological Sciences, Faculty of Science, Kuwait University, Safat, Kuwait
| | - Bincy Paul
- Biological Sciences, Faculty of Science, Kuwait University, Safat, Kuwait
| | - Divya Nair
- Biological Sciences, Faculty of Science, Kuwait University, Safat, Kuwait
| | - Jijin Kumar
- Department of Anatomy, Faculty of Medicine, Kuwait University, Safat, Kuwait
| | - Jassim M Al-Hassan
- Biological Sciences, Faculty of Science, Kuwait University, Safat, Kuwait
| |
Collapse
|
9
|
Ülger M, Sezer G, Özyazgan İ, Özocak H, Yay A, Balcıoğlu E, Yalçın B, Göç R, Ülger B, Özyazgan TM, Yakan B. The effect of erythropoietin and umbilical cord-derived mesenchymal stem cells on nerve regeneration in rats with sciatic nerve injury. J Chem Neuroanat 2021; 114:101958. [PMID: 33864937 DOI: 10.1016/j.jchemneu.2021.101958] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE We aimed to investigate the effects of umbilical cord-derived mesenchymal stem cells and erythropoietin on nerve regeneration in the sciatic nerve 'crush injury' in a rat model. METHODS Experimental animals were randomly divided into 5 groups: Crush Injury, Sham, Crush Injury + Erythropoietin, Crush Injury + Mesenchymal Stem Cell, Crush Injury + Erythropoietin + Mesenchymal Stem Cell groups. Crush injury made with bulldog clamp. Mesencyhmal stem cells delivered by enjection locally. Erythropoietin administered by intraperitoneally. On the 0th, 14th and 28th days, all groups underwent a sciatic functional index test. On 28th day, sciatic nerves were harvested and histopathological appearance, axon number and axon diameter of the sciatic nerves were evaluated with Oil Red O staining. Immunoreactivity of nerve growth factor, neurofilament-H and caspase-3 were determined by immunofluorescence staining in nerve tissue. RESULTS In histopathological examination, axons and nerve bundles exhibiting normal nerve architecture in the Sham group. Crush Injury + Mesenchymal Stem Cell group has similar histological appearance to the Sham group. The number of axons were higher in the Mesenchymal Stem Cell groups compared to the Crush Injury group. Nerve growth factor immunoreactivity intensity was significantly lower in Crush Injury + Mesenchymal Stem Cell group compared to Crush Injury group. Neurofilament-H density was higher in the treatment groups when compared to the Crush Injury group. CONCLUSIONS In this study, it was found that umbilical cord-derived mesenchymal stem cells and erythropoietin treatments effects positively regeneration of crush injury caused by bulldog clamp in the sciatic nerve of rats.
Collapse
Affiliation(s)
- Menekşe Ülger
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Gülay Sezer
- Department of Pharmacology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - İrfan Özyazgan
- Department of Plastic Reconstructive and Aesthetic Surgery, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Hakan Özocak
- Department of Plastic Reconstructive and Aesthetic Surgery, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Arzu Yay
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Esra Balcıoğlu
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Betül Yalçın
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Rümeysa Göç
- Department of Histology and Embryology, Cumhuriyet University, Faculty of Medicine, 058140, Sivas, Turkey.
| | - Birkan Ülger
- Department of Anesthesiology and Reanimation, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Tuğçe Merve Özyazgan
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| | - Birkan Yakan
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, 38039, Kayseri, Turkey.
| |
Collapse
|
10
|
Combined Use of Chitosan and Olfactory Mucosa Mesenchymal Stem/Stromal Cells to Promote Peripheral Nerve Regeneration In Vivo. Stem Cells Int 2021; 2021:6613029. [PMID: 33488738 PMCID: PMC7801080 DOI: 10.1155/2021/6613029] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/27/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022] Open
Abstract
Peripheral nerve injury remains a clinical challenge with severe physiological and functional consequences. Despite the existence of multiple possible therapeutic approaches, until now, there is no consensus regarding the advantages of each option or the best methodology in promoting nerve regeneration. Regenerative medicine is a promise to overcome this medical limitation, and in this work, chitosan nerve guide conduits and olfactory mucosa mesenchymal stem/stromal cells were applied in different therapeutic combinations to promote regeneration in sciatic nerves after neurotmesis injury. Over 20 weeks, the intervened animals were subjected to a regular functional assessment (determination of motor performance, nociception, and sciatic indexes), and after this period, they were evaluated kinematically and the sciatic nerves and cranial tibial muscles were evaluated stereologically and histomorphometrically, respectively. The results obtained allowed confirming the beneficial effects of using these therapeutic approaches. The use of chitosan NGCs and cells resulted in better motor performance, better sciatic indexes, and lower gait dysfunction after 20 weeks. The use of only NGGs demonstrated better nociceptive recoveries. The stereological evaluation of the sciatic nerve revealed identical values in the different parameters for all therapeutic groups. In the muscle histomorphometric evaluation, the groups treated with NGCs and cells showed results close to those of the group that received traditional sutures, the one with the best final values. The therapeutic combinations studied show promising outcomes and should be the target of new future works to overcome some irregularities found in the results and establish the combination of nerve guidance conduits and olfactory mucosa mesenchymal stem/stromal cells as viable options in the treatment of peripheral nerves after injury.
Collapse
|
11
|
Dong M, Shi B, Liu D, Liu JH, Zhao D, Yu ZH, Shen XQ, Gan JM, Shi BL, Qiu Y, Wang CC, Zhu ZZ, Shen QD. Conductive Hydrogel for a Photothermal-Responsive Stretchable Artificial Nerve and Coalescing with a Damaged Peripheral Nerve. ACS NANO 2020; 14:16565-16575. [PMID: 33025785 DOI: 10.1021/acsnano.0c05197] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Modern development of flexible electronics has made use of bioelectronic materials as artificial tissue in vivo. As hydrogels are more similar to nerve tissue, functional hydrogels have become a promising candidate for bioelectronics. Meanwhile, interfacing functional hydrogels and living tissues is at the forefront of bioelectronics. The peripheral nerve injury often leads to paralysis, chronic pain, neurologic disorders, and even disability, because it has affected the bioelectrical signal transmission between the brain and the rest of body. Here, a kind of light-stimuli-responsive and stretchable conducting polymer hydrogel (CPH) is developed to explore artificial nerve. The conductivity of CPH can be enhanced when illuminated by near-infrared light, which can promote the conduction of the bioelectrical signal. When CPH is mechanically elongated, it still has high durability of conductivity and, thus, can accommodate unexpected strain of nerve tissues in motion. Thereby, CPH can better serve as an implant of the serious peripheral nerve injury in vivo, especially in the case that the length of the missing nerve exceeds 10 mm.
Collapse
Affiliation(s)
- Mei Dong
- Department of Polymer Science and Engineering, Key Laboratory of High-Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, PR China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
- Jiangsu Provincial Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
| | - Bo Shi
- Department of Spine Surgery, Affiliated Drum Tower Hospital of Nanjing University, Nanjing, Jiangsu 210008, PR China
| | - Dun Liu
- Department of Spine Surgery, Affiliated Drum Tower Hospital of Nanjing University, Nanjing, Jiangsu 210008, PR China
| | - Jia-Hao Liu
- Department of Polymer Science and Engineering, Key Laboratory of High-Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, PR China
| | - Di Zhao
- Department of Polymer Science and Engineering, Key Laboratory of High-Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, PR China
| | - Zheng-Hang Yu
- Department of Polymer Science and Engineering, Key Laboratory of High-Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, PR China
| | - Xiao-Quan Shen
- Department of Polymer Science and Engineering, Key Laboratory of High-Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, PR China
| | - Jia-Min Gan
- Department of Polymer Science and Engineering, Key Laboratory of High-Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, PR China
| | - Ben-Long Shi
- Department of Spine Surgery, Affiliated Drum Tower Hospital of Nanjing University, Nanjing, Jiangsu 210008, PR China
| | - Yong Qiu
- Department of Spine Surgery, Affiliated Drum Tower Hospital of Nanjing University, Nanjing, Jiangsu 210008, PR China
| | - Chang-Chun Wang
- College of Material Science and Engineering, Nanjing Institute of Technology, Nanjing, Jiangsu 211167, PR China
- Jiangsu key laboratory of Advanced Structural Materials & Application Technology, Nanjing, Jiangsu 211167, PR China
| | - Ze-Zhang Zhu
- Department of Spine Surgery, Affiliated Drum Tower Hospital of Nanjing University, Nanjing, Jiangsu 210008, PR China
| | - Qun-Dong Shen
- Department of Polymer Science and Engineering, Key Laboratory of High-Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, PR China
| |
Collapse
|
12
|
Limonene, a food additive, and its active metabolite perillyl alcohol improve regeneration and attenuate neuropathic pain after peripheral nerve injury: Evidence for IL-1β, TNF-α, GAP, NGF and ERK involvement. Int Immunopharmacol 2020; 86:106766. [DOI: 10.1016/j.intimp.2020.106766] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022]
|
13
|
Tsuang FY, Chen MH, Lin FH, Yang MC, Liao CJ, Chang WH, Sun JS. Partial enzyme digestion facilitates regeneration of crushed nerve in rat. Transl Neurosci 2020; 11:251-263. [PMID: 33335765 PMCID: PMC7711954 DOI: 10.1515/tnsci-2020-0112] [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: 04/14/2020] [Revised: 05/23/2020] [Accepted: 06/12/2020] [Indexed: 11/15/2022] Open
Abstract
Peripheral nerve injury is a life-changing disability with significant socioeconomic consequences. In this rat model, we propose that partial enzyme digestion can facilitate the functional recovery of a crushed nerve. The sciatic nerves were harvested and in vitro cultured with the addition of Liberase to determine the appropriate enzyme amount in the hyaluronic acid (HA) membrane. Then, the sciatic nerve of adult male Sprague-Dawley rats was exposed, crushed, and then treated with partial enzyme digestion (either 0.001 or 0.002 unit/mm2 Liberase-HA membrane). The sciatic function index (SFI) for functional recovery of the sciatic nerve was evaluated. After 2 h of in vitro digestion, fascicles and axons were separated from each other, with the cells mobilized. Greater destruction of histology structures occurred in the high enzyme (Liberase-HA membrane at 0.002 unit/mm2) group at 24 h than in the low enzyme (0.001 unit/mm2) group at 48 h. In the SFI evaluation, the improvement in 0.001 unit/mm2 Liberase group was significantly better than control and 0.002 unit/mm2 Liberase group. Our study demonstrated that appropriate enzyme digestion had a significantly faster and earlier recovery.
Collapse
Affiliation(s)
- Fon-Yih Tsuang
- Institute of Biomedical Engineering, College of Engineering and College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Surgery, National Taiwan University Hospital, Taipei City, Taiwan
| | - Ming-Hong Chen
- Department of Surgery, Division of Neurosurgery, WanFang Hospital, Taipei, Taiwan
| | - Feng-Huei Lin
- Institute of Biomedical Engineering, College of Engineering and College of Medicine, National Taiwan University, Taipei, Taiwan.,Division of Medical Engineering, National Health Research Institute, Miaoli County, Taiwan
| | - Ming-Chia Yang
- Orthopedic Device Technology Division, Industrial Technology Research Institute, Hsinchu County, Taiwan
| | - Chun-Jen Liao
- Orthopedic Device Technology Division, Industrial Technology Research Institute, Hsinchu County, Taiwan
| | - Wen-Hsiang Chang
- Orthopedic Device Technology Division, Industrial Technology Research Institute, Hsinchu County, Taiwan
| | - Jui-Sheng Sun
- Department of Orthopedic Surgery, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
14
|
Lee HG, Choi JH, Jang YS, Kim UK, Kim GC, Hwang DS. Non-thermal plasma accelerates the healing process of peripheral nerve crush injury in rats. Int J Med Sci 2020; 17:1112-1120. [PMID: 32410841 PMCID: PMC7211154 DOI: 10.7150/ijms.44041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/13/2020] [Indexed: 01/10/2023] Open
Abstract
The objective of this study was to evaluate the effect of non-thermal plasma (NTP) on the healing process of peripheral nerve crush injuries, which can occur during dental implant procedures. For this, a rat model of sciatic nerve crush injury (SNCI) was adopted. The rats were divided into three groups: non-nerve damage (non-ND), nerve damage (ND), and ND+NTP group. To evaluate the sciatic nerve (SN) function, the static sciatic index was calculated, and the muscle and SN tissues were subjected to a histologic analysis. The results showed that NTP effectively accelerated the healing process of SNCI in rats. In contrast to the ND group, which showed approximately 60% recovery in the SN function, the NTP-treated rats showed complete recovery. Histologically, the NTP treatments not only accelerated the muscle healing, but also reduced the edema-like phenotype of the damaged SN tissues. In the ND group, the SN tissues had an accumulation of CD68-positive macrophages, partially destroyed axonal fibers and myelinated Schwann cells. Conversely, in the ND+NTP group, the macrophage accumulation was reduced and an overall regeneration of the damaged axon fibers and the myelin sheath was accomplished. The results of this study indicate that NTP can be used for healing of injured peripheral nerves.
Collapse
Affiliation(s)
- Hyeong-Geun Lee
- Department of Oral & Maxillofacial Surgery, School of Dentistry, Pusan National University
| | - Jeong-Hae Choi
- Department of Research and Development Center, FEAGLE Corporation, Yangsan, South Korea.,Department of Oral Anatomy and Cell Biology, School of Dentistry, Pusan National University, Yangsan, South Korea
| | - Yoon-Seo Jang
- Department of Oral & Maxillofacial Surgery, School of Dentistry, Pusan National University
| | - Uk-Kyu Kim
- Department of Oral & Maxillofacial Surgery, School of Dentistry, Pusan National University
| | - Gyoo-Cheon Kim
- Department of Research and Development Center, FEAGLE Corporation, Yangsan, South Korea.,Department of Oral Anatomy and Cell Biology, School of Dentistry, Pusan National University, Yangsan, South Korea
| | - Dae-Seok Hwang
- Department of Oral & Maxillofacial Surgery, School of Dentistry, Pusan National University
| |
Collapse
|
15
|
Functional Recovery Occurs Even After Partial Remyelination of Axon-Meshed Median and Ulnar Nerves in Mice. Neurochem Res 2019; 44:2230-2236. [DOI: 10.1007/s11064-019-02863-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 07/06/2019] [Accepted: 08/19/2019] [Indexed: 01/12/2023]
|
16
|
Bone Marrow Mesenchymal Stem Cell Transplantation Enhances Nerve Regeneration in a Rat Model of Hindlimb Replantation. Plast Reconstr Surg 2019; 143:758e-768e. [PMID: 30921125 DOI: 10.1097/prs.0000000000005412] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Successful limb replantation must be based not only on the viability of the amputated part but also on satisfactory long-term functional recovery. Once the vascular, skeletal, and soft-tissue problems have been taken care of, nerve recovery becomes the ultimate limiting factor. Unfortunately, nerve regeneration after limb replantation is impaired by several consequences. The authors tested the hypothesis that bone marrow mesenchymal stem cells could improve nerve regeneration outcomes in an experimental model of limb replantation. METHODS Twenty rats underwent replantation after total hindlimb amputation. Animals were subdivided into two groups: a replanted but nontreated control group and a replanted and bone marrow mesenchymal stem cell-transplanted group. Three months after surgery, nerve regeneration was assessed using functional, electrophysiologic, histomorphologic, and immunohistochemical analyses. RESULTS Bone marrow mesenchymal stem cell-treated animals showed significantly better sciatic functional index levels and higher compound muscle action potential amplitudes in comparison with the controls. Histomorphometric analysis revealed that the number of regenerating axons was approximately two-fold greater in the treated nerves. In addition, the mean g-ratio of these axons was within the optimal range. Immunohistochemical assessment revealed that expression of S-100 and myelin basic protein in the treated nerves was significantly higher than in controls. Correspondingly, the expression levels of anti-protein gene product 9.5 and vesicular acetylcholine transporter in motor endplates were also significantly higher. Finally, muscles in the bone marrow mesenchymal stem cell-transplanted group showed significantly larger average fiber areas. CONCLUSION The authors' findings demonstrate that it is possible to improve the degree of nerve regeneration after limb replantation by bone marrow mesenchymal stem cell transplantation.
Collapse
|
17
|
Bingham JR, Kniery KR, Jorstad NL, Horkayne-Szakaly I, Hoffer ZS, Salgar SK. "Stem cell therapy to promote limb function recovery in peripheral nerve damage in a rat model" - Experimental research. Ann Med Surg (Lond) 2019; 41:20-28. [PMID: 31011420 PMCID: PMC6463551 DOI: 10.1016/j.amsu.2019.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/26/2019] [Accepted: 03/24/2019] [Indexed: 02/06/2023] Open
Abstract
Background Optimizing nerve regeneration and mitigating muscle atrophy are the keys to successful outcomes in peripheral nerve damage. We investigated whether mesenchymal stem cell (MSC) therapy can improve limb function recovery in peripheral nerve damage. Materials and methods We used sciatic nerve transection/repair (SNR) and individual nerve transection/repair (INR; branches of sciatic nerve - tibial, peroneal, sural) models to study the effect of MSCs on proximal and distal peripheral nerve damages, respectively, in male Lewis rats. Syngeneic MSCs (5 × 106; passage≤6) or saline were administered locally and intravenously. Sensory/motor functions (SF/MF) of the limb were assessed. Results Rat MSCs (>90%) were CD29+, CD90+, CD34−, CD31− and multipotent. Total SF at two weeks post-SNR & INR with or without MSC therapy was ∼1.2 on a 0–3 grading scale (0 = No function; 3 = Normal); by 12 weeks it was 2.6–2.8 in all groups (n ≥ 9/group). MSCs accelerated SF onset. At eight weeks post-INR, sciatic function index (SFI), a measure of MF (0 = Normal; −100 = Nonfunctional) was −34 and −77 in MSC and vehicle groups, respectively (n ≥ 9); post-SNR it was −72 and −92 in MSC and vehicle groups, respectively. Long-term MF (24 weeks) was apparent in MSC treated INR (SFI -63) but not in SNR (SFI -100). Gastrocnemius muscle atrophy was significantly reduced (P < 0.05) in INR. Nerve histomorphometry revealed reduced axonal area (P < 0.01) but no difference in myelination (P > 0.05) in MSC treated INR compared to the naive contralateral nerve. Conclusion MSC therapy in peripheral nerve damage appears to improve nerve regeneration, mitigate flexion-contractures, and promote limb functional recovery. Mesenchymal stem cell (MSC) therapy improved limb functional recovery. MSCs improved nerve regeneration and mitigated foot flexion-contractures. Limb muscle atrophy was significantly reduced in individual nerve repair (INR). Functional recovery in distal nerve repair (INR) was superior to proximal (SNR). MSC therapy is attractive, feasible & promising in peripheral nerve injury repair.
Collapse
Affiliation(s)
- Jason R Bingham
- Department of Surgery, Madigan Army Medical Center, Tacoma, WA, 98431, USA
| | - Kevin R Kniery
- Department of Surgery, Madigan Army Medical Center, Tacoma, WA, 98431, USA
| | - Nikolas L Jorstad
- Department of Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Iren Horkayne-Szakaly
- Department of Neuropathology & Ophthalmic Pathology, Joint Pathology Center, Defense Health Agency, Silver Spring, MD, 20910, USA
| | - Zachary S Hoffer
- Department of Pathology, Madigan Army Medical Center, Tacoma, WA, 98431, USA
| | - Shashikumar K Salgar
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, 98431, USA
| |
Collapse
|
18
|
Rodríguez Sánchez DN, de Lima Resende LA, Boff Araujo Pinto G, de Carvalho Bovolato AL, Possebon FS, Deffune E, Amorim RM. Canine Adipose-Derived Mesenchymal Stromal Cells Enhance Neuroregeneration in a Rat Model of Sciatic Nerve Crush Injury. Cell Transplant 2019; 28:47-54. [PMID: 30369261 PMCID: PMC6322136 DOI: 10.1177/0963689718809045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/27/2018] [Accepted: 10/01/2018] [Indexed: 12/24/2022] Open
Abstract
Crush injuries in peripheral nerves are frequent and induce long-term disability with motor and sensory deficits. Due to axonal and myelin sheath disruptions, strategies for optimized axonal regeneration are needed. Multipotent mesenchymal stromal cells (MSC) are promising because of their anti-inflammatory properties and secretion of neurotrophins. The present study investigated the effect of canine adipose tissue MSC (Ad-MSC) transplantation in an experimental sciatic nerve crush injury. Wistar rats were divided into three groups: sham ( n = 8); Crush+PBS ( n = 8); Crush+MSC ( n = 8). Measurements of sciatic nerve functional index (SFI), muscle mass, and electromyography (EMG) were performed. Canine Ad-MSC showed mesodermal characteristics (CD34-, CD45-, CD44+, CD90+ and CD105+) and multipotentiality due to chondrogenic, adipogenic, and osteogenic differentiation. SFI during weeks 3 and 4 was significantly higher in the Crush+MSC group ( p < 0.001). During week 4, the EMG latency in the Crush+MSC groups had better near normality ( p < 0.05). The EMG amplitude showed results close to normality during week 4 in the Crush+MSC group ( p < 0.04). There were no statistical differences in muscle weight between the groups ( p > 0.05), but there was a tendency toward weight gain in the Crush+MSC groups. Better motor functional recovery after crush and perineural canine Ad-MSC transplantation was observed during week 2. This was maintained till week 4. In conclusion, the canine Ad-MSC transplantation showed early pro-regenerative effects between 2-4 weeks in the rat model of sciatic nerve crush injury.
Collapse
Affiliation(s)
- Diego Noé Rodríguez Sánchez
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), São Paulo, Brazil
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Luiz Antonio de Lima Resende
- Department of Neurology and Psychiatry, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Giovana Boff Araujo Pinto
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), São Paulo, Brazil
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Ana Lívia de Carvalho Bovolato
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Fábio Sossai Possebon
- Department of Veterinary Hygiene and Public Health, College of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), São Paulo, Brazil
| | - Elenice Deffune
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Rogério Martins Amorim
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), São Paulo, Brazil
| |
Collapse
|
19
|
Torul D, Bereket MC, Onger ME, Altun G. Comparison of the Regenerative Effects of Platelet-Rich Fibrin and Plasma Rich in Growth Factors on Injured Peripheral Nerve: An Experimental Study. J Oral Maxillofac Surg 2018; 76:1823.e1-1823.e12. [PMID: 29763577 DOI: 10.1016/j.joms.2018.04.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 03/30/2018] [Accepted: 04/10/2018] [Indexed: 11/30/2022]
Abstract
PURPOSE The aim of this study was to investigate the effects of platelet-rich fibrin (PRF) and plasma rich in growth factors (PRGF) on peripheral nerve injury in the early period of healing. MATERIAL AND METHODS Thirty Wistar albino rats were used in this study. Rats were divided into control (C), damaged (D), PRF, and PRGF groups. The left sciatic nerves of each group were identified as group C. Crush-type injury was performed on the right sciatic nerves of the D, PRF, and PRGF groups. In the PRF and PRGF groups, blood 2 mL was obtained to prepare the PRF and PRGF and the biomaterials were applied to the injured nerve area. After 8 weeks, functional, electrophysiologic, and stereological evaluations were performed. RESULTS For the electrophysiologic evaluation, the latency and amplitude values in the D, PRF, and PRGF groups were significantly lower than those in the C group (P > .05). According to the sciatic functional index result, there were significant differences between groups D and PRF and between groups D and PRGF (P = .000). For the stereological evaluations, although no significant difference was observed between the PRGF and C groups (P > .05), a significant difference was observed among the D, PRF, and PRGF groups for myelinated axon number. There were significant differences between groups D and PRF and between groups D and PRGF for axon area (P = .021 and .001, respectively). No significant difference was observed among the D, PRF, and PRGF groups for myelin sheath thickness and ratio of axon area to myelin sheath thickness (P > .05). CONCLUSIONS The results of this study suggest that PRGF increases nerve regeneration in the early period of healing and that the limited early action of PRF should be re-evaluated in the late period.
Collapse
Affiliation(s)
- Damla Torul
- Research Assistant, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Ondokuz Mayis University, Samsun, Turkey.
| | - Mehmet Cihan Bereket
- Associate Professor, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Ondokuz Mayis University, Samsun, Turkey
| | - Mehmet Emin Onger
- Assistant Professor, Department of Histology and Embryology, Faculty of Dentistry, Ondokuz Mayis University, Samsun, Turkey
| | - Gamze Altun
- Research Assistant, Department of Histology and Embryology, Faculty of Dentistry, Ondokuz Mayis University, Samsun, Turkey
| |
Collapse
|
20
|
Local low dose curcumin treatment improves functional recovery and remyelination in a rat model of sciatic nerve crush through inhibition of oxidative stress. Neuropharmacology 2018; 139:98-116. [PMID: 30018000 DOI: 10.1016/j.neuropharm.2018.07.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/16/2018] [Accepted: 07/02/2018] [Indexed: 12/12/2022]
Abstract
Traumatic injuries to peripheral nerves are frequent, however, specific pharmacological treatments are currently lacking. Curcumin has antioxidant, anti-inflammatory and neuroprotective properties but high oral doses are required for therapeutic use, particularly due to its low bioavailability. The aim of the present study was to investigate the effects of local and continuous treatment using low curcumin doses on functional recovery and nerve regeneration after rat sciatic nerve crush (SNC). Curcumin was administered by osmotic pumps with a catheter delivering the drug at the injury site (0.2 mg/day for 4 weeks). Functionally, early improvements in mechanical sensitivity, finger spacing of the injured paw, skilful walking and grip strength were observed in curcumin-treated animals. The curcumin treatment increased expression of compact myelin proteins (MPZ and PMP22), myelin sheath thickness and, correspondingly, increased motor and sensitive nerve conduction velocity. Microscopic analysis of gastrocnemius muscle indicated a curcumin-induced decrease in neurogenic lesions. Curcumin treatment reduced the production of reactive oxygen species (ROS) (which were notably produced by macrophages), lipid peroxidation and increased expression of transcription factor Nrf2. In silico analyses indicated that curcumin combines all the characteristics required to be an efficient lipid peroxidation inhibitor at the heart of biological membranes, hence protecting their degradation due to ROS. This antioxidant capacity is likely to contribute to the beneficial effects of curcumin after SNC injury. These results demonstrate that, when administrated locally, low doses of curcumin represent a promising therapy for peripheral nerve regeneration.
Collapse
|
21
|
Doyen PJ, Vergouts M, Pochet A, Desmet N, van Neerven S, Brook G, Hermans E. Inflammation-associated regulation of RGS in astrocytes and putative implication in neuropathic pain. J Neuroinflammation 2017; 14:209. [PMID: 29078779 PMCID: PMC5658970 DOI: 10.1186/s12974-017-0971-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 09/26/2017] [Indexed: 02/07/2023] Open
Abstract
Background Regulators of G-protein signaling (RGS) are major physiological modulators of G-protein-coupled receptors (GPCR) signaling. Several GPCRs expressed in both neurons and astrocytes participate in the central control of pain processing, and the reduced efficacy of analgesics in neuropathic pain conditions may rely on alterations in RGS function. The expression and the regulation of RGS in astrocytes is poorly documented, and we herein hypothesized that neuroinflammation which is commonly observed in neuropathic pain could influence RGS expression in astrocytes. Methods In a validated model of neuropathic pain, the spared nerve injury (SNI), the regulation of RGS2, RGS3, RGS4, and RGS7 messenger RNA (mRNA) was examined up to 3 weeks after the lesion. Changes in the expression of the same RGS were also studied in cultured astrocytes exposed to defined activation protocols or to inflammatory cytokines. Results We evidenced a differential regulation of these RGS in the lumbar spinal cord of animals undergoing SNI. In particular, RGS3 appeared upregulated at early stages after the lesion whereas expression of RGS2 and RGS4 was decreased at later stages. Decrease in RGS7 expression was already observed after 3 days and outlasted until 21 days after the lesion. In cultured astrocytes, we observed that changes in the culture conditions distinctly influenced the constitutive expression of these RGS. Also, brief exposures (4 to 8 h) to either interleukin-1β, interleukin-6, or tumor necrosis factor α caused rapid changes in the mRNA levels of the RGS, which however did not strictly recapitulate the regulations observed in the spinal cord of lesioned animals. Longer exposure (48 h) to inflammatory cytokines barely influenced RGS expression, confirming the rapid but transient regulation of these cell signaling modulators. Conclusion Changes in the environment of astrocytes mimicking the inflammation observed in the model of neuropathic pain can affect RGS expression. Considering the role of astrocytes in the onset and progression of neuropathic pain, we propose that the inflammation-mediated modulation of RGS in astrocytes constitutes an adaptive mechanism in a context of neuroinflammation and may participate in the regulation of nociception.
Collapse
Affiliation(s)
- Pierre J Doyen
- Neuropharmacology, Institute of Neuroscience, Université Catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Maxime Vergouts
- Neuropharmacology, Institute of Neuroscience, Université Catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Amandine Pochet
- Neuropharmacology, Institute of Neuroscience, Université Catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Nathalie Desmet
- Neuropharmacology, Institute of Neuroscience, Université Catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Sabien van Neerven
- Neuropharmacology, Institute of Neuroscience, Université Catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Gary Brook
- Institute for Neuropathology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Emmanuel Hermans
- Neuropharmacology, Institute of Neuroscience, Université Catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium.
| |
Collapse
|
22
|
Hussain ASM, Renno WM, Sadek HL, Kayali NM, Al-Salem A, Rao MS, Khan KM. Monoamine oxidase-B inhibitor protects degenerating spinal neurons, enhances nerve regeneration and functional recovery in sciatic nerve crush injury model. Neuropharmacology 2017; 128:231-243. [PMID: 29054367 DOI: 10.1016/j.neuropharm.2017.10.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/28/2017] [Accepted: 10/15/2017] [Indexed: 12/29/2022]
Abstract
Monoamine oxidase-B (MAOB), a flavin adenine dinucleotide (FAD), is an enzyme which catalyzes the oxidation of amines. MAOB is proposed to play a major role in the pathogenesis of neurodegeneration through the production of reactive oxygen species (ROS) and neurotoxins. The present study was designed to outline the effects of the MAOB inhibitor (MAOB-I) on neuroprotection of spinal neurons, regeneration of sciatic nerve fibers, and recovery of sensory-motor functions in the sciatic nerve crush injury model. Male Wistar rats (4-months-old) were assigned to i) Naïve (N), ii) Sham (S), iii) Sciatic nerve crush and treated with saline (CRUSH + SALINE) and iv) Sciatic nerve crush and treated with MAOB inhibitor (CRUSH + MAOB-I) groups (n = 10/group). In groups iii and iv, the crush injury was produced by crushing the sciatic nerve followed by treatment with saline or MAOB-I (Selegiline® 2.5 mg/kg) intraperitoneally for 10 days. Behavioral tests were conducted from week 1 to week 6. At the end of the study, sciatic nerve and lumbar spinal cord were examined by immunohistochemistry, light and electron microscopy. MAOB-I treatment showed significant improvement in sensory and motor functions compared to saline treatment (p < 0.05-0.001) in injured nerves. The morphological study showed a significantly increased number of nerve fibers in sciatic nerve distal to the site of injury (p < 0.05), with better myelination pattern in CRUSH + MAOB-I treated group compared to CRUSH + SALINE group. Spinal cord ventral horns showed a significant increase in the number of NeuN-immunoreactive neurons in the MAOB-I treated group compared to Saline treated group (p < 0.01). MAOB-I has a significant potential for protecting the degenerating spinal cord neurons and enhancing the regeneration of injured sciatic nerve fibers following crush injury.
Collapse
Affiliation(s)
| | - Waleed M Renno
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait.
| | - Hanaa L Sadek
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait
| | - Noura M Kayali
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait
| | - Aseel Al-Salem
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait
| | - Muddanna S Rao
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait
| | - Khalid M Khan
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait
| |
Collapse
|
23
|
Ozturk O, Tezcan AH, Adali Y, Yıldırım CH, Aksoy O, Yagmurdur H, Bilge A. Effect of ozone and methylprednisolone treatment following crush type sciatic nerve injury. Acta Cir Bras 2017; 31:730-735. [PMID: 27982260 DOI: 10.1590/s0102-865020160110000005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 10/17/2016] [Indexed: 12/19/2022] Open
Abstract
PURPOSE: To assess and compare the histopathological effects of ozone therapy and/or methylprednisolone (MPS) treatment on regeneration after crush type sciatic nerve injury. METHODS: Forty Sprague-Dawley male rats were randomly allocated into four groups. Four groups received the following regimens intraperitoneally every day for 14 days after formation of crush type injury on sciatic nerve: Group I: ozone (20mcg/ml); Group II: methylprednisolone (2mg/kg); Group III: ozone (20 mcg/ml) and methylprednisolone (2mg/kg); Group IV: isotonic saline (0.9%). The histomorphological evaluation was made after biopsies were obtained from the sites of injury. RESULTS: Significant differences were noted between groups in terms of degeneration (p=0.019), nerve sheath cell atrophy (p=0.012), intraneural inflammatory cellular infiltration (p=0.002), perineural granulation tissue formation (p=0.019), perineural vascular proliferation (p=0.004), perineural inflammatory cellular infiltration (p<0.001) and inflammation in peripheral tissue (p=0.006). Degeneration was remarkably low in Group III, while no change in nerve sheath cell was noted in Group II. CONCLUSION: The combined use of methylprednisolone and ozone treatment can have beneficial effects for regeneration after crush type nerve injury.
Collapse
Affiliation(s)
- Omur Ozturk
- Assistant Professor, Department of Anesthesiology and Reanimation, School of Medicine, Kafkas University, Kars, Turkey. Conception, design, scientific and intellectual contributions of the study, acquisition of data, statistical analysis, manuscript writing, final approval
| | - Aysu Hayriye Tezcan
- Assistant Professor, Department of Anesthesiology and Reanimation, School of Medicine, Kafkas University, Kars, Turkey. Conception, design, scientific and intellectual contributions of the study, acquisition of data, statistical analysis, manuscript writing, final approval
| | - Yasemen Adali
- Asstistant Professor, Department of Pathology, School of Medicine, Kafkas University, Kars, Turkey. Histopathological examinations
| | - Can Hakan Yıldırım
- Asstistant Professor, Department of Neurosurgery, School of Medicine, Kafkas University, Kars, Turkey. Analysis and interpretation of data
| | - Ozgur Aksoy
- Professor, Department of Surgery, School of Veterinary Medicine, Kafkas University, Kars, Turkey. Technical procedures
| | - Hatice Yagmurdur
- Professor, Department of Anesthesiology and Reanimation, School of Medicine, Kafkas University, Kars, Turkey. Critical revision, final approval
| | - Ali Bilge
- Assistant Professor, Department of Ortopedics and Traumatology, School of Medicine, Kafkas University, Kars, Turkey. Manuscript preparation, final approval
| |
Collapse
|
24
|
Hader M, Sporer ME, Roche AD, Unger E, Bergmeister KD, Wakolbinger R, Aszmann OC. Fascicular shifting: a novel technique to overcome large nerve defects. J Neurosurg Spine 2017; 27:723-731. [PMID: 28984513 DOI: 10.3171/2017.3.spine16276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Over the last decade, a number of authors have investigated the utility of different biological and synthetic matrices as alternatives to conventional nerve grafts. However, the autologous nerve graft remains the gold standard, even though it often involves using a pure sensory nerve to reconstruct a mixed or even a pure motor nerve. Furthermore, limited donor sites often necessitate a significant mismatch of needed nerve tissue, especially for large proximal nerve defects such as brachial plexus lesions. Here, the authors present a new technique that overcomes these problems: the fascicular shift procedure (FSP). A fascicular group of the nerve distal to the injury is harvested in a sufficient length to bridge the nerve defect. METHODS The method of fascicular shifting was tested at the sciatic nerve in 45 Lewis rats. In the experimental group, a 15-mm nerve defect was created and reconstructed with a fascicular group that was harvested directly distal to the gap. This group was compared with 1 negative control group (defect without reconstruction) and 3 positive control groups (sensory, motor, and mixed graft). After 12 weeks of nerve regeneration, outcome was evaluated using retrograde labeling, histomorphometric analysis, and muscle force analysis. RESULTS All reconstructed groups showed successful regeneration with various levels of function. The negative control group showed minimal force measurements that were of no functional value. The fascicular shift provided sufficient guidance to overcome nerve defects, had higher (p < 0.1) motor neuron counts (1958.75 ± 657.21) than the sensory graft (1263.50 ± 538.90), and was equal to motor grafts (1490.43 ± 794.80) and mixed grafts (1720.00 ± 866.421). This tendency of improved motor regeneration was confirmed in all analyses. The mixed graft group was compared with the experimental group to investigate the influence of the potential damage induced by the fascicular shift distal to the repair site. However, none of the analyses revealed an impairment of nerve regeneration for both the tibial and common peroneal index muscles. CONCLUSIONS This study demonstrates that harvesting a transplant from the nerve segment distal to the injury site offers a mixed graft without causing additional donor-site morbidity. These grafts perform statistically better than a standard sensory graft in terms of motor recovery. The fascicular shift presents a novel method to reconstruct large proximal nerve defects, making it immensely attractive in brachial plexus reconstruction.
Collapse
Affiliation(s)
- Maria Hader
- 1Christian Doppler Laboratory for Restoration of Extremity Function
| | | | - Aidan D Roche
- 1Christian Doppler Laboratory for Restoration of Extremity Function
| | - Ewald Unger
- 3Centre for Medical Physics and Biomedical Engineering, Medical University of Vienna, Austria
| | | | | | - Oskar C Aszmann
- 1Christian Doppler Laboratory for Restoration of Extremity Function.,2Division of Plastic and Reconstructive Surgery, Department of Surgery; and
| |
Collapse
|
25
|
Renno WM, Benov L, Khan KM. Possible role of antioxidative capacity of (-)-epigallocatechin-3-gallate treatment in morphological and neurobehavioral recovery after sciatic nerve crush injury. J Neurosurg Spine 2017; 27:593-613. [PMID: 28777065 DOI: 10.3171/2016.10.spine16218] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE This study examined the capacity of the major polyphenolic green tea extract (-)-epigallocatechin-3-gallate (EGCG) to suppress oxidative stress and stimulate the recovery and prompt the regeneration of sciatic nerve after crush injury. METHODS Adult male Wistar rats were randomly assigned to one of 4 groups: 1) Naïve, 2) Sham (sham injury, surgical control group), 3) Crush (sciatic nerve crush injury treated with saline), and 4) Crush+EGCG (sciatic nerve crush injury treated with intraperitoneally administered EGCG, 50 mg/kg). All animals were tested for motor and sensory neurobehavioral parameters throughout the study. Sciatic nerve and spinal cord tissues were harvested and processed for morphometric and stereological analysis. For the biochemical assays, the time points were Day 1, Day 7, Day 14, and Day 28 after nerve injury. RESULTS After sciatic nerve crush injury, the EGCG-treated animals (Crush+EGCG group) showed significantly better recovery of foot position and toe spread and 50% greater improvement in motor recovery than the saline-treated animals (Crush group). The Crush+EGCG group displayed an early hopping response at the beginning of the 3rd week postinjury. Animals in the Crush+EGCG group also showed a significant reduction in mechanical allodynia and hyperalgesia latencies and significant improvement in recovery from nociception deficits in both heat withdrawal and tail flick withdrawal latencies compared with the Crush group. In both the Crush+EGCG and Crush groups, quantitative evaluation revealed significant morphological evidence of neuroregeneration according to the following parameters: mean cross-sectional area of axons, myelin thickness in the sciatic nerve (from Week 4 to Week 8), increase of myelin basic protein concentration and gene expression in both the injured sciatic nerve and spinal cord, and fiber diameter to axon diameter ratio and myelin thickness to axon diameter ratio at Week 2 after sciatic nerve injury. However, the axon area remained much smaller in both the Crush+EGCG and Crush groups compared with the Sham and Naïve groups. The number of axons per unit area was significantly decreased in the Crush+EGCG and Crush groups compared with controls. Sciatic nerve injury produced generalized oxidative stress manifested as a significant increase of isoprostanes in the urine and decrease of the total antioxidant capacity (TAC) of the blood from Day 7 until Day 14. EGCG-treated rats showed significantly less increase of isoprostanes than saline-treated animals and also showed full recovery of TAC levels by Day 14 after nerve injury. In spinal cord tissue analysis, EGCG-treated animals showed induced glutathione reductase and suppressed induction of heme oxygenase 1 gene expression compared with nontreated animals. CONCLUSIONS EGCG treatment suppressed the crush-induced production of isoprostanes and stimulated the recovery of the TAC and was associated with remarkable alleviation of motor and sensory impairment and significant histomorphological evidence of neuronal regeneration following sciatic nerve crush injury in rats. The findings of this study suggest that EGCG can be used as an adjunctive therapeutic remedy for nerve injury. However, further investigations are needed to establish the antioxidative mechanism involved in the regenerative process after nerve injury. Only upregulation of glutathione reductase supports the idea that EGCG is acting indirectly via induction of enzymes or transcription factors.
Collapse
Affiliation(s)
| | - Ludmil Benov
- Biochemistry, Faculty of Medicine, Kuwait University, Safat, Kuwait
| | | |
Collapse
|
26
|
Huang Y, Bornstein MM, Lambrichts I, Yu HY, Politis C, Jacobs R. Platelet-rich plasma for regeneration of neural feedback pathways around dental implants: a concise review and outlook on future possibilities. Int J Oral Sci 2017; 9:1-9. [PMID: 28282030 PMCID: PMC5379164 DOI: 10.1038/ijos.2017.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2017] [Indexed: 02/05/2023] Open
Abstract
Along with the development of new materials, advanced medical imaging and surgical techniques, osseointegrated dental implants are considered a successful and constantly evolving treatment modality for the replacement of missing teeth in patients with complete or partial edentulism. The importance of restoring the peripheral neural feedback pathway and thus repairing the lack of periodontal mechanoreceptors after tooth extraction has been highlighted in the literature. Nevertheless, regenerating the nerve fibers and reconstructing the neural feedback pathways around osseointegrated implants remain a challenge. Recent studies have provided evidence that platelet-rich plasma (PRP) therapy is a promising treatment for musculoskeletal injuries. Because of its high biological safety, convenience and usability, PRP therapy has gradually gained popularity in the clinical field. Although much remains to be learned, the growth factors from PRP might play key roles in peripheral nerve repair mechanisms. This review presents known growth factors contributing to the biological efficacy of PRP and illustrates basic and (pre-)clinical evidence regarding the use of PRP and its relevant products in peripheral nerve regeneration. In addition, the potential of local application of PRP for structural and functional recovery of injured peripheral nerves around dental implants is discussed.
Collapse
Affiliation(s)
- Yan Huang
- OMFS IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven and Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium.,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Michael M Bornstein
- OMFS IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven and Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium.,Section of Dental Radiology and Stomatology, Department of Oral Surgery and Stomatology, University of Bern, Bern, Switzerland.,Applied Oral Sciences, Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, China
| | - Ivo Lambrichts
- Group of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Hai-Yang Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Constantinus Politis
- OMFS IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven and Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Reinhilde Jacobs
- OMFS IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven and Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
27
|
Kern B, Budihardjo JD, Mermulla S, Quan A, Cadmi C, Lopez J, Khusheim M, Xiang S, Park J, Furtmüller GJ, Sarhane KA, Schneeberger S, Lee WPA, Hoke A, Tuffaha SH, Brandacher G. A Novel Rodent Orthotopic Forelimb Transplantation Model That Allows for Reliable Assessment of Functional Recovery Resulting From Nerve Regeneration. Am J Transplant 2017; 17:622-634. [PMID: 27500557 DOI: 10.1111/ajt.14007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/28/2016] [Accepted: 08/04/2016] [Indexed: 01/25/2023]
Abstract
Improved nerve regeneration and functional outcomes would greatly enhance the utility of vascularized composite allotransplantation (VCA) such as hand and upper extremity transplantation. However, research aimed at achieving this goal has been limited by the lack of a functional VCA animal model. We have developed a novel rat midhumeral forelimb transplant model that allows for the characterization of upper extremity functional recovery following transplantation. At the final end point of 12 weeks, we found that animals with forelimb transplantation including median, ulnar and radial nerve coaptation demonstrated significantly improved grip strength and forelimb function as compared to forelimb transplantation without nerve approximation (grip strength: 1.71N ± 0.57 vs. no appreciable recovery; IBB scale: 2.6 ± 0.7? vs. 0.8 ± 0.40; p = 0.0005), and similar recovery to nerve transection-and-repair only (grip strength: 1.71N ± 0.57 vs. 2.03 ± 0.42.6; IBB scale: 2.6 ± 0.7 vs. 2.8 ± 0.8; p = ns). Moreover, all forelimb transplant animals with nerve coaptation displayed robust axonal regeneration with myelination and reduced flexor muscle atrophy when compared to forelimb transplant animals without nerve coaptation. In conclusion, this is the first VCA small-animal model that allows for reliable and reproducible measurement of behavioral functional recovery in addition to histologic evaluation of nerve regeneration and graft reinnervation.
Collapse
Affiliation(s)
- B Kern
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Visceral, Transplant, and Thoracic Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - J D Budihardjo
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD
| | - S Mermulla
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD
| | - A Quan
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD
| | - C Cadmi
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD
| | - J Lopez
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD
| | - M Khusheim
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD
| | - S Xiang
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD
| | - J Park
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
| | - G J Furtmüller
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD
| | - K A Sarhane
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD
| | - S Schneeberger
- Department of Visceral, Transplant, and Thoracic Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - W P A Lee
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD
| | - A Hoke
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
| | - S H Tuffaha
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD
| | - G Brandacher
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
28
|
Ribeiro J, Caseiro AR, Pereira T, Armada-da-Silva PA, Pires I, Prada J, Amorim I, Leal Reis I, Amado S, Santos JD, Bompasso S, Raimondo S, Varejão ASP, Geuna S, Luís AL, Maurício AC. Evaluation of PVA biodegradable electric conductive membranes for nerve regeneration in axonotmesis injuries: the rat sciatic nerve animal model. J Biomed Mater Res A 2017; 105:1267-1280. [PMID: 28078802 DOI: 10.1002/jbm.a.35998] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/13/2016] [Accepted: 01/05/2017] [Indexed: 11/06/2022]
Abstract
The therapeutic effect of three polyvinyl alcohol (PVA) membranes loaded with electrically conductive materials - carbon nanotubes (PVA-CNTs) and polypyrrole (PVA-PPy) - were tested in vivo for neuro-muscular regeneration after an axonotmesis injury in the rat sciatic nerve. The membranes electrical conductivity measured was 1.5 ± 0.5 × 10-6 S/m, 579 ± 0.6 × 10-6 S/m, and 1837.5 ± 0.7 × 10-6 S/m, respectively. At week-12, a residual motor and nociceptive deficit were present in all treated groups, but at week-12, a better recovery to normal gait pattern of the PVA-CNTs and PVA-PPy treated groups was observed. Morphometrical analysis demonstrated that PVA-CNTs group presented higher myelin thickness and lower g-ratio. The tibialis anterior muscle, in the PVA-PPy and PVA-CNTs groups showed a 9% and 19% increase of average fiber size area and a 5% and 10% increase of the "minimal Feret's diameter," respectively. No inflammation, degeneration, fibrosis or necrosis were detected in lung, liver, kidneys, spleen, and regional lymph nodes and absence of carbon deposits was confirmed with Von Kossa and Masson-Fontana stains. In conclusion, the membranes of PVA-CNTs and PVA-PPy are biocompatible and have electrical conductivity. The higher electrical conductivity measured in PVA-CNTs membrane might be responsible for the positive results on maturation of myelinated fibers. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 1267-1280, 2017.
Collapse
Affiliation(s)
- Jorge Ribeiro
- Departmento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Sub-inidade de Cirurgia Experimental e Medicina Regenerativa, Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal.,UPVET, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal
| | - Ana Rita Caseiro
- Departmento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Sub-inidade de Cirurgia Experimental e Medicina Regenerativa, Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal.,CEMUC, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, Porto, 4200-465, Portugal
| | - Tiago Pereira
- Departmento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Sub-inidade de Cirurgia Experimental e Medicina Regenerativa, Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
| | - Paulo Alexandre Armada-da-Silva
- Faculdade de Motricidade Humana (FMH), Universidade de Lisboa (ULisboa), Estrada da Costa, 1499-002, Dafundo, Cruz Quebrada, Portugal.,CIPER-FMH: Centro Interdisciplinar de Estudo de Performance Humana, Faculdade de Motricidade Humana (FMH), Universidade de Lisboa (ULisboa), Estrada da Costa, 1499-002, Cruz Quebrada - Dafundo, Portugal
| | - Isabel Pires
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal.,CECAV, Centro de Ciência Animal e Veterinária, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, Vila Real, 5000-801, Portugal
| | - Justina Prada
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal.,CECAV, Centro de Ciência Animal e Veterinária, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, Vila Real, 5000-801, Portugal
| | - Irina Amorim
- Departmento de Patologia e de Imunologia Molecular, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Rua Dr. Roberto Frias s/n, 4200-465, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto (UP), Rua Alfredo Allen, Porto, 4200-135, Portugal
| | - Inês Leal Reis
- Departmento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Sub-inidade de Cirurgia Experimental e Medicina Regenerativa, Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
| | - Sandra Amado
- Instituto Politécnico de Leiria, UIS-IPL: Unidade de Investigação em Saúde da Escola Superior de Saúde de Leiria, Portugal.,CDrsp - Centre for Rapid and Sustainable Product Development, Rua de Portugal 2430-028, Marinha, Grande, Portugal
| | - José Domingos Santos
- CEMUC, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, Porto, 4200-465, Portugal
| | - Simone Bompasso
- Department of Clinical and Biological Sciences, University of Turin, Turin, 10126, Italy.,Neuroscience Institute of the Cavalieri Ottolenghi Foundation (NICO), Azienda Ospedaliero-Universitaria San Luigi Gonzaga, Regione Gonzole 10, Orbassano, 10043, Turin, Italy
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences, University of Turin, Turin, 10126, Italy.,Neuroscience Institute of the Cavalieri Ottolenghi Foundation (NICO), Azienda Ospedaliero-Universitaria San Luigi Gonzaga, Regione Gonzole 10, Orbassano, 10043, Turin, Italy
| | - Artur Severo Proença Varejão
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal.,CECAV, Centro de Ciência Animal e Veterinária, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, Vila Real, 5000-801, Portugal
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, University of Turin, Turin, 10126, Italy.,Neuroscience Institute of the Cavalieri Ottolenghi Foundation (NICO), Azienda Ospedaliero-Universitaria San Luigi Gonzaga, Regione Gonzole 10, Orbassano, 10043, Turin, Italy
| | - Ana Lúcia Luís
- Departmento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Sub-inidade de Cirurgia Experimental e Medicina Regenerativa, Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal.,UPVET, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal
| | - Ana Colette Maurício
- Departmento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Sub-inidade de Cirurgia Experimental e Medicina Regenerativa, Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
| |
Collapse
|
29
|
|
30
|
Kuyucu E, Gümüs B, Erbas O, Oltulu F, Bora A. Exenatide promotes regeneration of injured rat sciatic nerve. Neural Regen Res 2017; 12:637-643. [PMID: 28553346 PMCID: PMC5436364 DOI: 10.4103/1673-5374.205105] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Damage to peripheral nerves results in partial or complete dysfunction. After peripheral nerve injuries, a full functional recovery usually cannot be achieved despite the standard surgical repairs. Neurotrophic factors and growth factors stimulate axonal growth and support the viability of nerve cells. The objective of this study is to investigate the neurotrophic effect of exenatide (glucagon like peptide-1 analog) in a rat sciatic nerve neurotmesis model. We injected 10 μg/d exenatide for 12 weeks in the experimental group (n = 12) and 0.1 mL/d saline for 12 weeks in the control group (n = 12). We evaluated nerve regeneration by conducting electrophysiological and motor functional tests. Histological changes were evaluated at weeks 1, 3, 6, and 9. Nerve regeneration was monitored using stereomicroscopy. The electrophysiological and motor functions in rats treated with exenatide were improved at 12 weeks after surgery. Histological examination revealed a significant increase in the number of axons in injured sciatic nerve following exenatide treatment confirmed by stereomicroscopy. In an experimentally induced neurotmesis model in rats, exenatide had a positive effect on nerve regeneration evidenced by electromyography, functional motor tests, histological and stereomicroscopic findings.
Collapse
Affiliation(s)
- Ersin Kuyucu
- Department of Orthopedics and Traumatology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Bilal Gümüs
- Orthopedics Clinic, Antalya State Hospital, Antalya, Turkey
| | - Oytun Erbas
- Department of Physiology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Fatih Oltulu
- Department of Histology Clinic, Ege University Faculty of Medicine, Izmir, Turkey
| | - Arslan Bora
- Department of Orthopedics and Traumatology, Izmir Atatürk Training and Educational Research Hospital, Izmir, Turkey
| |
Collapse
|
31
|
|
32
|
A new model of nerve injury in the rat reveals a role of Regulator of G protein Signaling 4 in tactile hypersensitivity. Exp Neurol 2016; 286:1-11. [PMID: 27641322 DOI: 10.1016/j.expneurol.2016.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 09/13/2016] [Accepted: 09/14/2016] [Indexed: 12/15/2022]
Abstract
Tactile hypersensitivity is one of the most debilitating symptoms of neuropathic pain syndromes. Clinical studies have suggested that its presence at early postoperative stages may predict chronic (neuropathic) pain after surgery. Currently available animal models are typically associated with consistent tactile hypersensitivity and are therefore limited to distinguish between mechanisms that underlie tactile hypersensitivity as opposed to mechanisms that protect against it. In this study we have modified the rat model of spared nerve injury, restricting the surgical lesion to a single peripheral branch of the sciatic nerve. This modification reduced the prevalence of tactile hypersensitivity from nearly 100% to approximately 50%. With this model, we here also demonstrated that the Regulator of G protein Signaling 4 (RGS4) was specifically up-regulated in the lumbar dorsal root ganglia and dorsal horn of rats developing tactile hypersensitivity. Intrathecal delivery of the RGS4 inhibitor CCG63802 was found to reverse tactile hypersensitivity for a 1h period. Moreover, tactile hypersensitivity after modified spared nerve injury was most frequently persistent for at least four weeks and associated with higher reactivity of glial cells in the lumbar dorsal horn. Based on these data we suggest that this new animal model of nerve injury represents an asset in understanding divergent neuropathic pain outcomes, so far unravelling a role of RGS4 in tactile hypersensitivity. Whether this model also holds promise in the study of the transition from acute to chronic pain will have to be seen in future investigations.
Collapse
|
33
|
He X, Ao Q, Wei Y, Song J. Transplantation of miRNA-34a overexpressing adipose-derived stem cell enhances rat nerve regeneration. Wound Repair Regen 2016; 24:542-50. [DOI: 10.1111/wrr.12427] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 01/17/2016] [Indexed: 01/10/2023]
Affiliation(s)
- Xingliang He
- Key Lab of School of Kinesiology; Shenyang Sport University and
- Department of Tissue Engeering; China Medical University; Shenyang Liaoning China
| | - Qiang Ao
- Department of Tissue Engeering; China Medical University; Shenyang Liaoning China
| | - Yujun Wei
- Department of Tissue Engeering; China Medical University; Shenyang Liaoning China
| | - Jinrui Song
- Key Lab of School of Kinesiology; Shenyang Sport University and
| |
Collapse
|
34
|
Renno WM, Khan KM, Benov L. Is there a role for neurotrophic factors and their receptors in augmenting the neuroprotective effect of (-)-epigallocatechin-3-gallate treatment of sciatic nerve crush injury? Neuropharmacology 2015; 102:1-20. [PMID: 26514400 DOI: 10.1016/j.neuropharm.2015.10.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/01/2015] [Accepted: 10/23/2015] [Indexed: 12/17/2022]
Abstract
This study analyzed and compared the effects of EGCG treatment on the expression of NTFs and NTF receptors expression in the sciatic nerve and the L3-L6 spinal cord segments at the early phase of regeneration following sciatic nerve crush injury. Analysis of BDNF, GDNF and NT3 neurotropic factors and Trk-B, Trk-C and NGFR-p75 receptors in neurons in the spinal cord of CRUSH and CRUSH + EGGC rats showed significant (p < 0.0001) decrease compared to NAÏVE and SHAM at day 1, 3, 7 and 14 after nerve injury. EGCG treatment significantly (p < 0.0001) increased the BDNF, GDN, NT3, Trk-B, Trk-C and NGFR-p75 immunostaining in the L3-L6 spinal cord compared to CRUSH animals. Also, EGCG treatment significantly increased the Trk-B protein concentration and Trk-B, NT3 and Trk-C gene expression in the spinal cords compared to CRUSH group. However, at day 1 and 3 post nerve injury, EGCG treatment significantly decreased the NGFR-p75 expression compared to CRUSH rats. In the sciatic nerve, EGCG treatment significantly (p < 0.01) increased the Trk-B and NGFR-p75 protein concentration in the controls. EGCG treatment significantly (p < 0.0001) increased the Trk-B, Trk-C and NGFR-p75 mRNA gene expressions in the sciatic nerves compared to CRUSH group. Only at day 1, CRUSH + EGCG animals displayed significant rise in the sciatic nerves NT3 gene expression compared to CRUSH group. Our data suggest that the EGCG neuroprotective effect on the spinal cord neurons may be mediated through the modulation of NTFs and NTF receptors following nerve crush injury in a rat model.
Collapse
Affiliation(s)
- Waleed M Renno
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait.
| | - Khalid M Khan
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait
| | - Ludmil Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait
| |
Collapse
|
35
|
A systematic review of animal models for experimental neuroma. J Plast Reconstr Aesthet Surg 2015; 68:1447-63. [DOI: 10.1016/j.bjps.2015.05.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 05/11/2015] [Accepted: 05/18/2015] [Indexed: 01/06/2023]
|
36
|
Ribeiro J, Pereira T, Caseiro AR, Armada-da-Silva P, Pires I, Prada J, Amorim I, Amado S, França M, Gonçalves C, Lopes MA, Santos JD, Silva DM, Geuna S, Luís AL, Maurício AC. Evaluation of biodegradable electric conductive tube-guides and mesenchymal stem cells. World J Stem Cells 2015; 7:956-975. [PMID: 26240682 PMCID: PMC4515438 DOI: 10.4252/wjsc.v7.i6.956] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/19/2015] [Accepted: 05/06/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the therapeutic effect of three tube-guides with electrical conductivity associated to mesenchymal stem cells (MSCs) on neuro-muscular regeneration after neurotmesis.
METHODS: Rats with 10-mm gap nerve injury were tested using polyvinyl alcohol (PVA), PVA-carbon nanotubes (CNTs) and MSCs, and PVA-polypyrrole (PPy). The regenerated nerves and tibialis anterior muscles were processed for stereological studies after 20 wk. The functional recovery was assessed serially for gait biomechanical analysis, by extensor postural thrust, sciatic functional index and static sciatic functional index (SSI), and by withdrawal reflex latency (WRL). In vitro studies included cytocompatibility, flow cytometry, reverse transcriptase polymerase chain reaction and karyotype analysis of the MSCs. Histopathology of lung, liver, kidneys, and regional lymph nodes ensured the biomaterials biocompatibility.
RESULTS: SSI remained negative throughout and independently from treatment. Differences between treted groups in the severity of changes in WRL existed, showing a faster regeneration for PVA-CNTs-MSCs (P < 0.05). At toe-off, less acute ankle joint angles were seen for PVA-CNTs-MSCs group (P = 0.051) suggesting improved ankle muscles function during the push off phase of the gait cycle. In PVA-PPy and PVA-CNTs groups, there was a 25% and 42% increase of average fiber area and a 13% and 21% increase of the “minimal Feret’s diameter” respectively. Stereological analysis disclosed a significantly (P < 0.05) increased myelin thickness (M), ratio myelin thickness/axon diameter (M/d) and ratio axon diameter/fiber diameter (d/D; g-ratio) in PVA-CNT-MSCs group (P < 0.05).
CONCLUSION: Results revealed that treatment with MSCs and PVA-CNTs tube-guides induced better nerve fiber regeneration. Functional and kinematics analysis revealed positive synergistic effects brought by MSCs and PVA-CNTs. The PVA-CNTs and PVA-PPy are promising scaffolds with electric conductive properties, bio- and cytocompatible that might prevent the secondary neurogenic muscular atrophy by improving the reestablishment of the neuro-muscular junction.
Collapse
|
37
|
An Experimental Comparison of the Effects of Propolis, Curcumin, and Methylprednisolone on Crush Injuries of the Sciatic Nerve. Ann Plast Surg 2015; 74:684-92. [DOI: 10.1097/sap.0000000000000026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
38
|
Jang SH, Lee JH. Effects of physical exercise on the functional recovery of rat hindlimbs with impairments of the sciatic nerve as assessed by 2D video analysis. J Phys Ther Sci 2015; 27:935-8. [PMID: 25931763 PMCID: PMC4395747 DOI: 10.1589/jpts.27.935] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 11/19/2014] [Indexed: 11/24/2022] Open
Abstract
[Purpose] The purpose of this study was to investigate the effects of treadmill training
on functional recovery by analyzing the ankle joint as well as the knee and hip joints
with 2D video analysis during gait by rats with sciatic nerve injury. [Subjects and
Methods] Twenty-four male Sprague-Dawley rats were used in this study. The sham group (SG)
received only a sham operation without any sciatic injury; the training group (TG)
performed treadmill training for 4 weeks after sciatic injury; and the control group (CG)
wasn’t provided with any therapeutic intervention after sciatic injury. [Results] The
ankle, knee, and hip ROM of TG and CG during the initial, mid stance, and toe-off phases
of gait at post-test were significantly different from SG. [Conclusion] Physical exercise,
like treadmill training, is beneficial for the improvement of the ankle, knee and hip
joints of rats with crushed sciatic nerve injury.
Collapse
Affiliation(s)
- Sang-Hun Jang
- Department of Physical Therapy, Gimcheon University, Republic of Korea
| | - Jung-Ho Lee
- Department of Physical Therapy, School of Medical and Public Health, Kyungdong University, Republic of Korea
| |
Collapse
|
39
|
Wen J, Sun D, Tan J, Young W. A consistent, quantifiable, and graded rat lumbosacral spinal cord injury model. J Neurotrauma 2015; 32:875-92. [PMID: 25313633 PMCID: PMC4492780 DOI: 10.1089/neu.2013.3321] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The purpose of this study is to develop a rat lumbosacral spinal cord injury (SCI) model that causes consistent motoneuronal loss and behavior deficits. Most SCI models focus on the thoracic or cervical spinal cord. Lumbosacral SCI accounts for about one third of human SCI but no standardized lumbosacral model is available for evaluating therapies. Twenty-six adult female Sprague-Dawley rats were randomized to three groups: sham (n=9), 25 mm (n=8), and 50 mm (n=9). Sham rats had laminectomy only, while 25 mm and 50 mm rats were injured by dropping a 10 g rod from a height of 25 mm or 50 mm, respectively, onto the L4-5 spinal cord at the T13/L1 vertebral junction. We measured footprint length (FL), toe spreading (TS), intermediate toe spreading (ITS), and sciatic function index (SFI) from walking footprints, and static toe spreading (STS), static intermediate toe spreading (SITS), and static sciatic index (SSI) from standing footprints. At six weeks, we assessed neuronal and white matter loss, quantified axons, diameter, and myelin thickness in the peroneal and tibial nerves, and measured cross-sectional areas of tibialis anterior and gastrocnemius muscle fibers. The result shows that peroneal and tibial motoneurons were respectively distributed in 4.71 mm and 5.01 mm columns in the spinal cord. Dropping a 10-g weight from 25 mm or 50 mm caused 1.5 mm or 3.75 mm gaps in peroneal and tibial motoneuronal columns, respectively, and increased spinal cord white matter loss. Fifty millimeter contusions significantly increased FL and reduced TS, ITS, STS, SITS, SFI, and SSI more than 25 mm contusions, and resulted in smaller axon and myelinated axon diameters in tibial and peroneal nerves and greater atrophy of gastrocnemius and anterior tibialis muscles, than 25 mm contusions. This model of lumbosacral SCI produces consistent and graded loss of white matter, motoneuronal loss, peripheral nerve axonal changes, and anterior tibialis and gastrocnemius muscles atrophy in rats.
Collapse
Affiliation(s)
- Junxiang Wen
- 1 Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey , Piscataway, New Jersey.,2 Department of Orthopaedics, Tongji University School of Medicine , Shanghai, China
| | - Dongming Sun
- 1 Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey , Piscataway, New Jersey
| | - Jun Tan
- 2 Department of Orthopaedics, Tongji University School of Medicine , Shanghai, China
| | - Wise Young
- 1 Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey , Piscataway, New Jersey
| |
Collapse
|
40
|
Karimi H, Forootan KS, Moein G, Mosavi SJ, Iekta BG. Survey of the results of acute sciatic nerve repair comparing epineural and perineurial techniques in the lower extremities of rat. JOURNAL OF ACUTE DISEASE 2015. [DOI: 10.1016/s2221-6189(14)60076-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
41
|
Gallo A, Dimiziani A, Damblon J, Michot B, Des Rieux A, De Kock M, Hermans E, Deumens R. Modulation of spinal glial reactivity by intrathecal PPF is not sufficient to inhibit mechanical allodynia induced by nerve crush. Neurosci Res 2015; 95:78-82. [PMID: 25697394 DOI: 10.1016/j.neures.2015.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/29/2015] [Accepted: 02/07/2015] [Indexed: 12/16/2022]
Abstract
Spinal glial reactivity has been strongly implicated in pain that follows peripheral nerve injury. Among the many therapeutic agents that have been tested for anti-allodynia through immune modulation is the atypical methylxanthine propentofylline. While propentofylline shows a potent anti-allodynia effect after nerve transection injury, we here demonstrate that, when propentofylline is used intrathecally at the effective immune-modulatory dose, allodynia after rat nerve crush injury is completely preserved. Microglial/macrophage Iba-1 and astrocytic GFAP expression, increased in the dorsal horn of nerve crushed animals, was, however, effectively attenuated by propentofylline. Effective modulation of spinal glial reactivity is, thus, no assurance for anti-allodynia.
Collapse
Affiliation(s)
- Alessandro Gallo
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200 Brussels, Belgium
| | - Andrea Dimiziani
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200 Brussels, Belgium
| | - Jonathan Damblon
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200 Brussels, Belgium
| | - Benoît Michot
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200 Brussels, Belgium
| | - Anne Des Rieux
- Louvain Drug Research Institute, Pharmaceutics and Drug Delivery Unit, Avenue E. Mounier 73, 1200 Brussels, Belgium
| | - Marc De Kock
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200 Brussels, Belgium
| | - Emmanuel Hermans
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200 Brussels, Belgium
| | - Ronald Deumens
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200 Brussels, Belgium.
| |
Collapse
|
42
|
Gärtner A, Pereira T, Simões MJ, Armada-da-Silva PA, França ML, Sousa R, Bompasso S, Raimondo S, Shirosaki Y, Nakamura Y, Hayakawa S, Osakah A, Porto B, Luís AL, Varejão AS, Maurício AC. Use of hybrid chitosan membranes and human mesenchymal stem cells from the Wharton jelly of umbilical cord for promoting nerve regeneration in an axonotmesis rat model. Neural Regen Res 2014; 7:2247-58. [PMID: 25538746 PMCID: PMC4268725 DOI: 10.3969/j.issn.1673-5374.2012.29.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 07/10/2012] [Indexed: 12/11/2022] Open
Abstract
Many studies have been dedicated to the development of scaffolds for improving post-traumatic nerve regeneration. The goal of this study was to assess the effect on nerve regeneration, associating a hybrid chitosan membrane with non-differentiated human mesenchymal stem cells isolated from Wharton's jelly of umbilical cord, in peripheral nerve reconstruction after crush injury. Chromosome analysis on human mesenchymal stem cell line from Wharton's jelly was carried out and no structural alterations were found in metaphase. Chitosan membranes were previously tested in vitro, to assess their ability in supporting human mesenchymal stem cell survival, expansion, and differentiation. For the in vivo testing, Sasco Sprague adult rats were divided in 4 groups of 6 or 7 animals each: Group 1, sciatic axonotmesis injury without any other intervention (Group 1-Crush); Group 2, the axonotmesis lesion of 3 mm was infiltrated with a suspension of 1 250–1 500 human mesenchymal stem cells (total volume of 50 μL) (Group 2-CrushCell); Group 3, axonotmesis lesion of 3 mm was enwrapped with a chitosan type III membrane covered with a monolayer of non-differentiated human mesenchymal stem cells (Group 3-CrushChitIIICell) and Group 4, axonotmesis lesion of 3 mm was enwrapped with a chitosan type III membrane (Group 4-CrushChitIII). Motor and sensory functional recovery was evaluated throughout a healing period of 12 weeks using sciatic functional index, static sciatic index, extensor postural thrust, and withdrawal reflex latency. Stereological analysis was carried out on regenerated nerve fibers. Results showed that infiltration of human mesenchymal stem cells, or the combination of chitosan membrane enwrapment and human mesenchymal stem cell enrichment after nerve crush injury provide a slight advantage to post-traumatic nerve regeneration. Results obtained with chitosan type III membrane alone confirmed that they significantly improve post-traumatic axonal regrowth and may represent a very promising clinical tool in peripheral nerve reconstructive surgery. Yet, umbilical cord human mesenchymal stem cells, that can be expanded in culture and induced to form several different types of cells, may prove, in future experiments, to be a new source of cells for cell therapy, including targets such as peripheral nerve and muscle.
Collapse
Affiliation(s)
- Andrea Gärtner
- Animal Science and Study Centre / Food and Agrarian Sciences and Technologies Institute, Porto University, 4099-003 Porto, Portugal ; Institute of Biomedical Sciences Abel Salazar, Veterinary Clinics Department, Porto University, Porto 4099-003, Portugal
| | - Tiago Pereira
- Animal Science and Study Centre / Food and Agrarian Sciences and Technologies Institute, Porto University, 4099-003 Porto, Portugal ; Institute of Biomedical Sciences Abel Salazar, Veterinary Clinics Department, Porto University, Porto 4099-003, Portugal
| | - Maria João Simões
- Animal Science and Study Centre / Food and Agrarian Sciences and Technologies Institute, Porto University, 4099-003 Porto, Portugal ; Institute of Biomedical Sciences Abel Salazar, Veterinary Clinics Department, Porto University, Porto 4099-003, Portugal
| | - Paulo As Armada-da-Silva
- Faculty of Human Kinetics, Technical University of Lisbon, Cruz Quebrada - Dafundo, 1499-002, Portugal
| | - Miguel L França
- Animal Science and Study Centre / Food and Agrarian Sciences and Technologies Institute, Porto University, 4099-003 Porto, Portugal ; Institute of Biomedical Sciences Abel Salazar, Veterinary Clinics Department, Porto University, Porto 4099-003, Portugal
| | - Rosa Sousa
- Institute of Biomedical Sciences Abel Salazar, Cytogenetic Department, Porto University, Porto 4099-003, Portugal
| | - Simone Bompasso
- Neuroscience Institute of the Cavalieri Ottolenghi Foundation, Orbassano 10043, Turin, Italy ; Department of Clinical and Biological Sciences, University of Turin, Orbassano 10010, Turin, Italy
| | - Stefania Raimondo
- Neuroscience Institute of the Cavalieri Ottolenghi Foundation, Orbassano 10043, Turin, Italy ; Department of Clinical and Biological Sciences, University of Turin, Orbassano 10010, Turin, Italy
| | - Yuki Shirosaki
- Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Yuri Nakamura
- Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Satoshi Hayakawa
- Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Akiyoshi Osakah
- Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Beatriz Porto
- Institute of Biomedical Sciences Abel Salazar, Cytogenetic Department, Porto University, Porto 4099-003, Portugal
| | - Ana Lúcia Luís
- Animal Science and Study Centre / Food and Agrarian Sciences and Technologies Institute, Porto University, 4099-003 Porto, Portugal ; Institute of Biomedical Sciences Abel Salazar, Veterinary Clinics Department, Porto University, Porto 4099-003, Portugal
| | - Artur Sp Varejão
- Department of Veterinary Sciences, Research Centre in Sports, Health and Human Development, University of Trás-os-Montes and Alto Douro, Vila Real 5001-801, Portugal
| | - Ana Colette Maurício
- Animal Science and Study Centre / Food and Agrarian Sciences and Technologies Institute, Porto University, 4099-003 Porto, Portugal ; Institute of Biomedical Sciences Abel Salazar, Veterinary Clinics Department, Porto University, Porto 4099-003, Portugal
| |
Collapse
|
43
|
Periodical assessment of electrophysiological recovery following sciatic nerve crush via surface stimulation in rats. Neurol Sci 2014; 36:449-56. [DOI: 10.1007/s10072-014-2005-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/04/2014] [Indexed: 01/31/2023]
|
44
|
Renno WM, Al-Maghrebi M, Rao MS, Khraishah H. (-)-Epigallocatechin-3-gallate modulates spinal cord neuronal degeneration by enhancing growth-associated protein 43, B-cell lymphoma 2, and decreasing B-cell lymphoma 2-associated x protein expression after sciatic nerve crush injury. J Neurotrauma 2014; 32:170-84. [PMID: 25025489 DOI: 10.1089/neu.2014.3491] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Our previous studies have established that (-)-epigallocatechin-3-gallate (EGCG) has both neuroprotective and -regenerative capacity after sciatic nerve injury. Moreover, this improvement was evident on the behavioral level. The aim of this study was to investigate the central effects of ECGC on spinal cord motor neurons after sciatic nerve injury. Our study showed that administering 50 mg/kg intraperitoneally i.p. of EGCG to sciatic nerve-injured rats improved their performance on different motor functions and mechanical hyperesthesia neurobehavioral tests. Histological analysis of spinal cords of EGCG-treated sciatic nerve-injured (CRUSH+ECGC) animals showed an increase in the number of neurons in the anterior horn, when compared to the naïve, sham, and saline-treated sciatic nerve-injured (CRUSH) control groups. Additionally, immunohistochemical study of spinal cord sections revealed that EGCG reduced the expression of glial fibrillary acidic protein and increased the expression of growth-associated protein 43, a marker of regenerating axons. Finally, EGCG reduced the ratio of B-cell lymphoma 2 (Bcl-2)-associated X protein/Bcl-2 and increased the expression of survivin gene. This study may shed some light on the future clinical use of EGCG and its constituents in the treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Waleed M Renno
- 1 Department of Anatomy, Faculty of Medicine, Kuwait University , Safat, Kuwait
| | | | | | | |
Collapse
|
45
|
Wang CZ, Chen YJ, Wang YH, Yeh ML, Huang MH, Ho ML, Liang JI, Chen CH. Low-level laser irradiation improves functional recovery and nerve regeneration in sciatic nerve crush rat injury model. PLoS One 2014; 9:e103348. [PMID: 25119457 PMCID: PMC4131879 DOI: 10.1371/journal.pone.0103348] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 06/27/2014] [Indexed: 12/21/2022] Open
Abstract
The development of noninvasive approaches to facilitate the regeneration of post-traumatic nerve injury is important for clinical rehabilitation. In this study, we investigated the effective dose of noninvasive 808-nm low-level laser therapy (LLLT) on sciatic nerve crush rat injury model. Thirty-six male Sprague Dawley rats were divided into 6 experimental groups: a normal group with or without 808-nm LLLT at 8 J/cm2 and a sciatic nerve crush injury group with or without 808-nm LLLT at 3, 8 or 15 J/cm2. Rats were given consecutive transcutaneous LLLT at the crush site and sacrificed 20 days after the crush injury. Functional assessments of nerve regeneration were analyzed using the sciatic functional index (SFI) and hindlimb range of motion (ROM). Nerve regeneration was investigated by measuring the myelin sheath thickness of the sciatic nerve using transmission electron microscopy (TEM) and by analyzing the expression of growth-associated protein 43 (GAP43) in sciatic nerve using western blot and immunofluorescence staining. We found that sciatic-injured rats that were irradiated with LLLT at both 3 and 8 J/cm2 had significantly improved SFI but that a significant improvement of ROM was only found in rats with LLLT at 8 J/cm2. Furthermore, the myelin sheath thickness and GAP43 expression levels were significantly enhanced in sciatic nerve-crushed rats receiving 808-nm LLLT at 3 and 8 J/cm2. Taken together, these results suggest that 808-nm LLLT at a low energy density (3 J/cm2 and 8 J/cm2) is capable of enhancing sciatic nerve regeneration following a crush injury.
Collapse
Affiliation(s)
- Chau-Zen Wang
- Department of Physiology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Jen Chen
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yan-Hsiung Wang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Long Yeh
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Mao-Hsiung Huang
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Ling Ho
- Department of Physiology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jen-I Liang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Hsin Chen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail:
| |
Collapse
|
46
|
The mouse median nerve experimental model in regenerative research. BIOMED RESEARCH INTERNATIONAL 2014; 2014:701682. [PMID: 25180190 PMCID: PMC4142669 DOI: 10.1155/2014/701682] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 07/23/2014] [Accepted: 07/25/2014] [Indexed: 01/12/2023]
Abstract
Sciatic nerve crush injury in rat animal model is one of the most common experimental models used in regenerative research. However, the availability of transgenic mouse for nerve regeneration studies is constantly increasing and, therefore, the shift from rat model to mouse model is, in some cases, necessary. Moreover, since most of the human nerve lesions occur in the upper limb, it is also advantageous to shift from sciatic nerve to median nerve. In this study we described an experimental model which involves lesions of the median nerve in the mouse. Data showed that the finger flexor muscle contraction strength, assessed to evaluate the motor function recovery, and reached values not different from the control already 20 days after injury. The degree of nerve regeneration evaluated with stereological methods in light microscopy showed that, 25 days after injury, the number of regenerated myelinated fibers was comparable to the control, but they were smaller with a thinner myelin thickness. Stereological analysis made in electron microscopy confirmed these results, although the total number of fibers quantified was significantly higher compared to light microscopy analysis, due to the very small size of some fibers that can be detected only in electron microscopy.
Collapse
|
47
|
Wark HAC, Mathews KS, Normann RA, Fernandez E. Behavioral and cellular consequences of high-electrode count Utah Arrays chronically implanted in rat sciatic nerve. J Neural Eng 2014; 11:046027. [DOI: 10.1088/1741-2560/11/4/046027] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
48
|
Challenges for nerve repair using chitosan-siloxane hybrid porous scaffolds. BIOMED RESEARCH INTERNATIONAL 2014; 2014:153808. [PMID: 25054129 PMCID: PMC4087280 DOI: 10.1155/2014/153808] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/29/2014] [Accepted: 05/31/2014] [Indexed: 01/02/2023]
Abstract
The treatment of peripheral nerve injuries remains one of the greatest challenges of neurosurgery, as functional recover is rarely satisfactory in these patients. Recently, biodegradable nerve guides have shown great potential for enhancing nerve regeneration. A major advantage of these nerve guides is that no foreign material remains after the device has fulfilled its task, which spares a second surgical intervention. Recently, we studied peripheral nerve regeneration using chitosan-γ-glycidoxypropyltrimethoxysilane (chitosan-GPTMS) porous hybrid membranes. In our studies, these porous membranes significantly improved nerve fiber regeneration and functional recovery in rat models of axonotmetic and neurotmetic sciatic nerve injuries. In particular, the number of regenerated myelinated nerve fibers and myelin thickness were significantly higher in rat treated with chitosan porous hybrid membranes, whether or not they were used in combination with mesenchymal stem cells isolated from the Wharton's jelly of the umbilical cord. In this review, we describe our findings on the use of chitosan-GPTMS hybrids for nerve regeneration.
Collapse
|
49
|
Ribeiro J, Gartner A, Pereira T, Gomes R, Lopes MA, Gonçalves C, Varejão A, Luís AL, Maurício AC. Perspectives of employing mesenchymal stem cells from the Wharton's jelly of the umbilical cord for peripheral nerve repair. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 108:79-120. [PMID: 24083432 DOI: 10.1016/b978-0-12-410499-0.00004-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mesenchymal stem cells (MSCs) from Wharton's jelly present high plasticity and low immunogenicity, turning them into a desirable form of cell therapy for the injured nervous system. Their isolation, expansion, and characterization have been performed from cryopreserved umbilical cord tissue. Great concern has been dedicated to the collection, preservation, and transport protocols of the umbilical cord after the parturition to the laboratory in order to obtain samples with higher number of viable MSCs without microbiological contamination. Different biomaterials like chitosan-silicate hybrid, collagen, PLGA90:10, poly(DL-lactide-ɛ-caprolactone), and poly(vinyl alcohol) loaded with electrical conductive materials, associated to MSCs have also been tested in the rat sciatic nerve in axonotmesis and neurotmesis lesions. The in vitro studies of the scaffolds included citocompatibility evaluation of the biomaterials used and cell characterization by imunocytochemistry, karyotype analysis, differentiation capacity into neuroglial-like cells, and flow cytometry. The regeneration process follow-up has been performed by functional analysis and the repaired nerves processed for stereological studies permitted the morphologic regeneration evaluation. The MSCs from Wharton's jelly delivered through tested biomaterials should be regarded a potentially valuable tool to improve clinical outcome especially after trauma to sensory nerves. In addition, these cells represent a noncontroversial source of primitive mesenchymal progenitor cells, which can be harvested after birth, cryogenically stored, thawed, and expanded for therapeutic uses. The importance of a longitudinal study concerning tissue engineering of the peripheral nerve, which includes a multidisciplinary team able to develop biomaterials associated to cell therapies, to perform preclinical trials concerning animal welfare and the appropriate animal model is here enhanced.
Collapse
Affiliation(s)
- Jorge Ribeiro
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal; Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências e Tecnologias Agrárias e Agro-Alimentares (ICETA), Universidade do Porto (UP), Porto, Portugal
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kakegawa A, Yokouchi K, Itsubo T, Kawagishi K, Karasawa M, Moriizumi T, Fukushima N. Correlation between motor function and axonal morphology in neonatally sciatic nerve-injured rats. Anat Sci Int 2014; 90:97-103. [PMID: 24771539 DOI: 10.1007/s12565-014-0236-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 04/09/2014] [Indexed: 12/22/2022]
Abstract
The present study was conducted to investigate the correlation between motor function and axonal morphology in neonatally sciatic nerve-injured rats. The left sciatic nerve of newborn rats was transected or crushed, and functionality of the sciatic nerve was assessed by the static sciatic index after 8 weeks. After functional assessment, the common peroneal nerves in the control, nerve-transected, and nerve-crushed rats were removed and prepared for morphometric examinations. The cross-sectional area of the nerve, total number of myelinated axons, and size of each myelinated axon were analyzed for each group. The control rats showed normal motor function, whereas the nerve-transected rats showed severe motor dysfunction. The cross-sectional area of the nerve and total number of myelinated axons were reduced after nerve transection. Moreover, the percentage per size class of myelinated axons was almost uniform in the control rats, while the distribution was shifted to the left in the nerve-transected rats. Furthermore, no large myelinated axons were observed in the nerve-transected rats. The nerve-crushed rats showed various gait functions with various distribution patterns of axonal size, and the rats were divided into two groups with and without uninjured residual large axons. The results showed that the importance of regenerated medium-sized axons in cases without large axons and of residual large axons in cases with large axons in motor function. It was revealed that motor function was related closely to axonal size in neonatally nerve-injured rats.
Collapse
Affiliation(s)
- Akira Kakegawa
- Department of Anatomy, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | | | | | | | | | | | | |
Collapse
|