1
|
State of the Art of Hydrogel Wound Dressings Developed by Ionizing Radiation. Gels 2023; 9:gels9010055. [PMID: 36661821 PMCID: PMC9858288 DOI: 10.3390/gels9010055] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
The development of an ideal hydrogel wound dressing with excellent characteristics is currently a significant demand in wound therapy. The ideal hydrogel wound dressing must provide a moist environment between the wound and the dressing, promote wound healing, absorb excess exudate and toxins, be completely sterile, and not adhere to the wound. The evolution and current status of research on hydrogel wound dressings obtained exclusively through production by ionizing radiation are discussed in this paper review, along with the preparation methods, properties, standard characterization techniques, and their applications in wound dressing. First, we described the methods for synthesizing hydrogel wound dressings with ionizing radiation. Then, standard methods of characterization of hydrogel wound dressings such as gel fraction, swelling degree, sol-gel analysis, rheological properties, morphology, moisture retention capability, and water vapor transmission rate have been investigated. In the end, specific attention was paid to the drug release, antibacterial performance, and cytotoxicity of hydrogels. Moreover, the application of hydrogel in regenerative medicine as wound healing dressing was covered.
Collapse
|
2
|
Zhang D, Su Y, Sun P, Liu X, Zhang L, Ling X, Fan Y, Wu K, Shi Q, Liu J. A TGF-loading hydrogel scaffold capable of promoting chondrogenic differentiation for repairing rabbit nasal septum cartilage defect. Front Bioeng Biotechnol 2022; 10:1057904. [DOI: 10.3389/fbioe.2022.1057904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/24/2022] [Indexed: 11/21/2022] Open
Abstract
Hydrogel-based tissue engineering has been widely used to repair cartilage injury. However, whether this approach can be applied to treat nasal septum cartilage defects remains unclear. In this study, three gelatin methacrylate-based scaffolds loaded with transforming growth factor (TGF)-β1 (GelMA-T) were prepared, and their effects on repair of nasal septum cartilage defects were examined. In vitro, the GelMA-T scaffolds showed good biocompatibility and promoted the chondrogenic differentiation of bone mesenchymal stem cells. Among three scaffolds, the 10% GelMA-T scaffold promoted chondrogenic differentiation most effectively, which significantly improved the expression of chondrocyte-related genes, including Col II, Sox9, and ACAN. In vivo, 10% GelMA-T scaffolds and 10% GelMA-T scaffolds loaded with bone mesenchymal stem cells (BMSCs; 10% GelMA-T/BMSCs) were transplanted into a nasal septum cartilage defect site in a rabbit model. At 4, 12, and 24 weeks after surgery, the nasal septum cartilage defects exhibited more complete repair in rabbits treated with the 10% GelMA-T/BMSC scaffold as demonstrated by hematoxylin & eosin, safranine-O, and toluidine blue staining. We showed that GelMA-T/BMSCs can be applied in physiological and structural repair of defects in nasal septum cartilage, providing a potential strategy for repairing cartilage defects in the clinic.
Collapse
|
3
|
Qiu G, Huang M, Ke D, Liu J, Weir MD, Ma T, Wang P, Oates TW, Schneider A, Xia Y, Xu HHK, Zhao L. Novel injectable calcium phosphate scaffold with human periodontal ligament stem cell encapsulation in microbeads for bone regeneration. FRONTIERS IN MATERIALS 2022; 9. [DOI: 10.3389/fmats.2022.977853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Objectives: 1) Develop a novel construct of human periodontal ligament stem cells (hPDLSCs) encapsulated in degradable alginate microbeads (DAMB) with human platelet lysate (hPL) and injectable calcium phosphate cement (ICPC); 2) Investigate the proliferation and osteogenic differentiation of hPDLSCs in ICPC with hPL as a xeno-free supplement and animal serum replacement for bone tissue engineering applications.Methods: hPDLSCs were encapsulated in alginate-fibrin microbeads (DAMB + fibrin), alginate-hPL degradable microbeads (DAMB + hPL), or alginate-fibrin-hPL microbeads (DAMB + fibrin + hPL). The proliferation and osteogenic differentiation of hPDLSCs were investigated in culturing with the ICPC scaffold.Results: Flexural strength of ICPC was 8.4 ± 0.91 MPa, and elastic modulus was 1.56 ± 0.1 GPa, exceeding those of cancellous bone. hPDLSCs had higher viability in DAMB + fibrin + hPL group than in DAMB + fibrin. ALP was 69.97 ± 16.96 mU/mg for ICPC + DAMB + fibrin + hPL group, higher than 30.68 ± 2.86 mU/mg of ICPC + DAMB + fibrin (p < 0.05) and 4.12 ± 1.65 mU/mg of control (p < 0.01). At 7 days, osteogenic gene expressions (ALP, RUNX2, COL1, and OPN) in ICPC + DAMB + fibrin + hPL and ICPC + DAMB + fibrin were 4–11 folds that of control. At 21 days, the hPDLSC-synthesized bone mineral amounts in ICPC + DAMB + fibrin + hPL and ICPC + DAMB + fibrin were 13.2 folds and 11.1 folds that of control group, respectively.Conclusion: The novel injectable CPC scaffold encapsulating hPDLSCs and hPL is promising to protect and deliver hPDLSCs. The hPL-based medium significantly enhanced the osteogenic differentiation of hPDLSCs in ICPC + DAMB + fibrin + hPL construct, suggesting a promising xeno-free approach for bone tissue regeneration applications.
Collapse
|
4
|
Qiu G, Huang M, Liu J, Ma T, Schneider A, Oates TW, Lynch CD, Weir MD, Zhang K, Zhao L, Xu HHK. Human periodontal ligament stem cell encapsulation in alginate-fibrin-platelet lysate microbeads for dental and craniofacial regeneration. J Dent 2022; 124:104219. [PMID: 35817226 DOI: 10.1016/j.jdent.2022.104219] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Tissue engineering is promising for dental and craniofacial regeneration. The objectives of this study were to develop a novel xeno-free alginate-fibrin-platelet lysate hydrogel with human periodontal ligament stem cells (hPDLSCs) for dental regeneration, and to investigate the proliferation and osteogenic differentiation of hPDLSCs using hPL as a cell culture nutrient supplement. METHODS hPDLSCs were cultured with Dulbecco's modified eagle medium (DMEM), DMEM + 10% fetal bovine serum (FBS), and DMEM + hPL (1%, 2.5%, and 5%). hPDLSCs were encapsulated in alginate-fibrin microbeads (Alg+Fib), alginate-hPL microbeads (Alg+hPL), or alginate-fibrin-hPL microbeads (Alg+Fib+hPL). hPDLSCs encapsulated in alginate microbeads were induced with an osteogenic medium containing hPL or FBS. Quantitative real-time polymerase chain reaction (qRT-PCR), alkaline phosphatase (ALP) activity, ALP staining, and alizarin red (ARS) staining was investigated. RESULTS hPDLSCs were released faster from Alg+Fib+hPL than from Alg+hPL. At 14 days, ALP activity was 44.1 ± 7.61 mU/mg for Alg+Fib+hPL group, higher than 28.07 ± 5.15 mU/mg of Alg+Fib (p<0.05) and 0.95 ± 0.2 mU/mg of control (p<0.01). At 7 days, osteogenic genes (ALP, RUNX2, COL1, and OPN) in Alg+Fib+hPL and Alg+Fib were 3-10 folds those of control. At 21 days, the hPDLSC-synthesized bone mineral amount in Alg+Fib+hPL and Alg+Fib was 7.5 folds and 4.3 folds that of control group, respectively. CONCLUSIONS The 2.5% hPL was determined to be optimal for hPDLSCs. Adding hPL into alginate hydrogel improved the viability of the hPDLSCs encapsulated in the microbeads. The hPL-based medium enhanced the osteogenic differentiation of hPDLSCs in Alg+Fib+hPL construct, showing a promising xeno-free approach for delivering hPDLSCs to enhance dental, craniofacial and orthopedic regenerations.
Collapse
Affiliation(s)
- Gengtao Qiu
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China; Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, United States of America; Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingguang Huang
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Jin Liu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, United States of America; Key Laboratory of Shannxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shannxi, China
| | - Tao Ma
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, United States of America
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, United States of America; Member, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| | - Thomas W Oates
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, United States of America
| | - Christopher D Lynch
- Restorative Dentistry, University Dental School and Hospital, University College Cork, Wilton, Cork, Ireland
| | - Michael D Weir
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, United States of America.
| | - Ke Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China.
| | - Liang Zhao
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China; Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Hockin H K Xu
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China; Member, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America; Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States of America
| |
Collapse
|
5
|
Deng X, Gould M, Ali MA. A review of current advancements for wound healing: Biomaterial applications and medical devices. J Biomed Mater Res B Appl Biomater 2022; 110:2542-2573. [PMID: 35579269 PMCID: PMC9544096 DOI: 10.1002/jbm.b.35086] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 12/12/2022]
Abstract
Wound healing is a complex process that is critical in restoring the skin's barrier function. This process can be interrupted by numerous diseases resulting in chronic wounds that represent a major medical burden. Such wounds fail to follow the stages of healing and are often complicated by a pro‐inflammatory milieu attributed to increased proteinases, hypoxia, and bacterial accumulation. The comprehensive treatment of chronic wounds is still regarded as a significant unmet medical need due to the complex symptoms caused by the metabolic disorder of the wound microenvironment. As a result, several advanced medical devices, such as wound dressings, wearable wound monitors, negative pressure wound therapy devices, and surgical sutures, have been developed to correct the chronic wound environment and achieve skin tissue regeneration. Most medical devices encompass a wide range of products containing natural (e.g., chitosan, keratin, casein, collagen, hyaluronic acid, alginate, and silk fibroin) and synthetic (e.g., polyvinyl alcohol, polyethylene glycol, poly[lactic‐co‐glycolic acid], polycaprolactone, polylactic acid) polymers, as well as bioactive molecules (e.g., chemical drugs, silver, growth factors, stem cells, and plant compounds). This review addresses these medical devices with a focus on biomaterials and applications, aiming to deliver a critical theoretical reference for further research on chronic wound healing.
Collapse
Affiliation(s)
- Xiaoxuan Deng
- Centre for Bioengineering & Nanomedicine (Dunedin), Department of Oral Rehabilitation, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Maree Gould
- Centre for Bioengineering & Nanomedicine (Dunedin), Department of Oral Rehabilitation, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - M Azam Ali
- Centre for Bioengineering & Nanomedicine (Dunedin), Department of Oral Rehabilitation, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
6
|
Ladeira BMF, Gomes MC, Custódio CA, Mano JF. High-Throughput Production of Microsponges from Platelet Lysate for Tissue Engineering Applications. Tissue Eng Part C Methods 2022; 28:325-334. [PMID: 35343236 DOI: 10.1089/ten.tec.2022.0029] [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: 11/12/2022] Open
Abstract
Cell-based therapies require a large number of cells, as well as appropriate methods to deliver the cells to damaged tissue. Microcarriers provide an optimal platform for large-scale cell culture while also improving cell retention during cell delivery. However, this technology still presents significant challenges due to low-throughput fabrication methods and an inability of the microcarriers to recreate the properties of human tissue. This work proposes, for the first time, the use of methacryloyl platelet lysates (PLMA), a photocrosslinkable material derived from human platelet lysates, to produce porous microcarriers. Initially, high quantities of PLMA/alginate core-shell microcapsules are produced using coaxial electrospray. Subsequently, the microcapsules are collected, irradiated with ultraviolet light, washed, and freeze dried yielding PLMA microsponges. These microsponges are able to support the adhesion and proliferation of human adipose-derived stem cells, while also displaying potential in the assembly of autologous microtissues. Cell-laden microsponges were shown to self-organize into aggregates, suggesting possible applications in bottom-up tissue engineering applications. Impact Statement Microcarriers have increasingly been used as delivery platforms in cell therapy. Herein, the encapsulation of human-derived proteins in alginate microcapsules is proposed as a method to produce microcarriers from photopolymerizable materials. The capsules function as a template structure, which is then processed into spherical microparticles, which can be used in cell culture, cell delivery, and bottom-up assembly. As a proof of concept, this method was combined with lyophilization to process methacryloyl platelet lysates into injectable microsponges for cell delivery.
Collapse
Affiliation(s)
- Bruno M F Ladeira
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Maria C Gomes
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Catarina A Custódio
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
7
|
Menter DG, Afshar-Kharghan V, Shen JP, Martch SL, Maitra A, Kopetz S, Honn KV, Sood AK. Of vascular defense, hemostasis, cancer, and platelet biology: an evolutionary perspective. Cancer Metastasis Rev 2022; 41:147-172. [PMID: 35022962 PMCID: PMC8754476 DOI: 10.1007/s10555-022-10019-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/04/2022] [Indexed: 01/08/2023]
Abstract
We have established considerable expertise in studying the role of platelets in cancer biology. From this expertise, we were keen to recognize the numerous venous-, arterial-, microvascular-, and macrovascular thrombotic events and immunologic disorders are caused by severe, acute-respiratory-syndrome coronavirus 2 (SARS-CoV-2) infections. With this offering, we explore the evolutionary connections that place platelets at the center of hemostasis, immunity, and adaptive phylogeny. Coevolutionary changes have also occurred in vertebrate viruses and their vertebrate hosts that reflect their respective evolutionary interactions. As mammals adapted from aquatic to terrestrial life and the heavy blood loss associated with placentalization-based live birth, platelets evolved phylogenetically from thrombocytes toward higher megakaryocyte-blebbing-based production rates and the lack of nuclei. With no nuclei and robust RNA synthesis, this adaptation may have influenced viral replication to become less efficient after virus particles are engulfed. Human platelets express numerous receptors that bind viral particles, which developed from archetypal origins to initiate aggregation and exocytic-release of thrombo-, immuno-, angiogenic-, growth-, and repair-stimulatory granule contents. Whether by direct, evolutionary, selective pressure, or not, these responses may help to contain virus spread, attract immune cells for eradication, and stimulate angiogenesis, growth, and wound repair after viral damage. Because mammalian and marsupial platelets became smaller and more plate-like their biophysical properties improved in function, which facilitated distribution near vessel walls in fluid-shear fields. This adaptation increased the probability that platelets could then interact with and engulf shedding virus particles. Platelets also generate circulating microvesicles that increase membrane surface-area encounters and mark viral targets. In order to match virus-production rates, billions of platelets are generated and turned over per day to continually provide active defenses and adaptation to suppress the spectrum of evolving threats like SARS-CoV-2.
Collapse
Affiliation(s)
- David G Menter
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - John Paul Shen
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie L Martch
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth V Honn
- Department of Pathology, Bioactive Lipids Research Program, Wayne State University, 5101 Cass Ave. 430 Chemistry, Detroit, MI, 48202, USA
- Department of Pathology, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
- Cancer Biology Division, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
8
|
Berni P, Leonardi F, Conti V, Ramoni R, Grolli S, Mattioli G. Case Report: A Novel Ventilated Thermoplastic Mesh Bandage for Post-operative Management of Large Soft Tissue Defects: A Case Series of Three Dogs Treated With Autologous Platelet Concentrates. Front Vet Sci 2021; 8:704567. [PMID: 34540933 PMCID: PMC8440817 DOI: 10.3389/fvets.2021.704567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/12/2021] [Indexed: 11/24/2022] Open
Abstract
A ventilated thermoplastic mesh bandage was used for the post-operative management of large soft tissue defects in three dogs. Once the granulation tissue appeared, the wounds were treated with liquid or jellified autologous platelet concentrates, Platelet Rich Plasma (PRP) and Platelet Lysate (PL), to improve the wound healing process. After cleaning the wound with sterile physiological solution, a dressing was performed with several layers of cotton. A window through the layers of cotton was opened above the wound. Then, the platelet concentrate was topically applied, and the bandage was completed by placing, over the access window, a ventilated thermoplastic mesh modeled according to the size and shape of the wound. After 24 h, it was replaced by a low adhesion bandage. The thermoplastic mesh avoids the direct contact between the wound and the external layers of the bandage, preventing the drainage of the topical agent and the removal of the growing healthy granulation tissue. The bandage proposed in this study is easily applied by the veterinarian and well-tolerated by the animal, ensuring high welfare standards in stressed patients presenting compromised clinical conditions.
Collapse
Affiliation(s)
- Priscilla Berni
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Fabio Leonardi
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Virna Conti
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Roberto Ramoni
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Stefano Grolli
- Department of Veterinary Science, University of Parma, Parma, Italy
| | | |
Collapse
|
9
|
Bonferoni MC, Caramella C, Catenacci L, Conti B, Dorati R, Ferrari F, Genta I, Modena T, Perteghella S, Rossi S, Sandri G, Sorrenti M, Torre ML, Tripodo G. Biomaterials for Soft Tissue Repair and Regeneration: A Focus on Italian Research in the Field. Pharmaceutics 2021; 13:pharmaceutics13091341. [PMID: 34575417 PMCID: PMC8471088 DOI: 10.3390/pharmaceutics13091341] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 12/22/2022] Open
Abstract
Tissue repair and regeneration is an interdisciplinary field focusing on developing bioactive substitutes aimed at restoring pristine functions of damaged, diseased tissues. Biomaterials, intended as those materials compatible with living tissues after in vivo administration, play a pivotal role in this area and they have been successfully studied and developed for several years. Namely, the researches focus on improving bio-inert biomaterials that well integrate in living tissues with no or minimal tissue response, or bioactive materials that influence biological response, stimulating new tissue re-growth. This review aims to gather and introduce, in the context of Italian scientific community, cutting-edge advancements in biomaterial science applied to tissue repair and regeneration. After introducing tissue repair and regeneration, the review focuses on biodegradable and biocompatible biomaterials such as collagen, polysaccharides, silk proteins, polyesters and their derivatives, characterized by the most promising outputs in biomedical science. Attention is pointed out also to those biomaterials exerting peculiar activities, e.g., antibacterial. The regulatory frame applied to pre-clinical and early clinical studies is also outlined by distinguishing between Advanced Therapy Medicinal Products and Medical Devices.
Collapse
Affiliation(s)
| | | | | | - Bice Conti
- Correspondence: (M.C.B.); (B.C.); (F.F.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Jeyaraman M, Muthu S, Khanna M, Jain R, Anudeep TC, Muthukanagaraj P, Siddesh SE, Gulati A, Satish AS, Jeyaraman N, Khanna V. Platelet lysate for COVID-19 pneumonia-a newer adjunctive therapeutic avenue. Stem Cell Investig 2021; 8:11. [PMID: 34268440 PMCID: PMC8256133 DOI: 10.21037/sci-2020-042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 04/27/2021] [Indexed: 02/05/2023]
Abstract
The linchpin for COVID-19 pathogenesis is the severe inflammatory process in the respiratory tract wherein the accumulation of excessive cytokines paves the way for a series of systemic hemodynamic alterations and mortality. The mortality rate is higher in individuals with co-morbidities and advancing age. The absence of a specific therapy is responsible for this uncontrolled spread and the significant mortality. This renders potential insight for considering biologics as a plausible option to repair and regenerate the affected lung tissue and pulverize the causative organism. The plausible role of megakaryocytes against invading microbes was not clearly understood. Platelet lysate is an acellular product consisting of regenerative molecules released from a cluster of platelets. It attenuates the changes caused by immune reactions in allogenic utility with the introduction of growth factors, cytokines, and proteins at supraphysiologic levels and thereby serves as a regenerative immunomodulatory agent to combat COVID-19. This platelet lysate can be used in nebulized form for such acute respiratory distress conditions in COVID-19 elderly patients. Platelet lysate may emerge as a pivotal player provided investigations pace up in this context. Here, we discuss how the platelet lysate can plausibly perquisite to relegate COVID-19. Undertaking prospective randomized controlled trials to prove its efficacy is the need of the hour in this pandemic scenario.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Orthopedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Sathish Muthu
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Orthopedics, Government Medical College & Hospital, Dindigul, Tamil Nadu, India
| | - Manish Khanna
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Orthopedics, Prasad Institute of Medical Science and Hospital, Lucknow, Uttar Pradesh, India
| | - Rashmi Jain
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Talagavadi Channaiah Anudeep
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Plastic Surgery, Topiwala National Medical College and BYL Nair Ch. Hospital, Mumbai, Maharashtra, India
| | - Purushothaman Muthukanagaraj
- Department of Internal Medicine & Psychiatry, SUNY-Upstate Binghamton Clinical Campus, Binghamton, New York, USA
| | | | - Arun Gulati
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Orthopedics, Kalpana Chawla Government Medical College & Hospital, Karnal, Haryana, India
| | | | - Naveen Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Orthopedics, Kasturba Medical College, MAHE Unievrsity, Manipal, Karnataka, India
| | - Venus Khanna
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Pathology, Prasad Institute of Medical Science and Hospital, Lucknow, Uttar Pradesh, India
| |
Collapse
|
11
|
Lech W, Sarnowska A, Kuczynska Z, Dabrowski F, Figiel-Dabrowska A, Domanska-Janik K, Buzanska L, Zychowicz M. Biomimetic microenvironmental preconditioning enhance neuroprotective properties of human mesenchymal stem cells derived from Wharton's Jelly (WJ-MSCs). Sci Rep 2020; 10:16946. [PMID: 33037314 PMCID: PMC7547118 DOI: 10.1038/s41598-020-74066-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023] Open
Abstract
Tuning stem cells microenvironment in vitro may influence their regenerative properties. In this study Wharton's Jelly-derived mesenchymal stem cells (WJ-MSCs) were encapsulated in 3D hydrogels derived from human fibrin (FB) or platelet lysate (PL) and the oxygen level was adjusted to physiological normoxia (5% O2). The influence of the type of the scaffold and physiological normoxia conditions was tested on the WJ-MSCs' survivability, proliferation, migratory potential, the level of expression of selected trophic factors, cytokines, and neural markers. Encapsulated WJ-MSCs revealed high survivability, stable proliferation rate, and ability to migrate out of the hydrogel and the up-regulated expression of all tested factors, as well as the increased expression of neural differentiation markers. Physiological normoxia stimulated proliferation of encapsulated WJ-MSCs and significantly enhanced their neuronal, but not glial, differentiation. Ex vivo studies with indirect co-culture of organotypic hippocampal slices and cell-hydrogel bio-constructs revealed strong neuroprotective effect of WJ-MSCs against neuronal death in the CA1 region of the rat hippocampus. This effect was potentiated further by FB scaffolds under 5% O2 conditions. Our results indicating significant effect of oxygen and 3D cytoarchitecture suggest the urgent need for further optimization of the microenvironmental conditions to improve therapeutical competence of the WJ-MSCs population.
Collapse
Affiliation(s)
- Wioletta Lech
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106, Warsaw, Poland
| | - Anna Sarnowska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106, Warsaw, Poland.,Translational Platform for Regenerative Medicine, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106, Warsaw, Poland
| | - Zuzanna Kuczynska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106, Warsaw, Poland
| | - Filip Dabrowski
- 1st Department of Obstetrics and Gynecology, Faculty of Medicine, Medical University of Warsaw, Starynkiewicza Square 1/3, 02-015, Warsaw, Poland
| | - Anna Figiel-Dabrowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106, Warsaw, Poland
| | - Krystyna Domanska-Janik
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106, Warsaw, Poland
| | - Leonora Buzanska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106, Warsaw, Poland
| | - Marzena Zychowicz
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106, Warsaw, Poland.
| |
Collapse
|
12
|
Jahangir S, Eglin D, Pötter N, Khozaei Ravari M, Stoddart MJ, Samadikuchaksaraei A, Alini M, Baghaban Eslaminejad M, Safa M. Inhibition of hypertrophy and improving chondrocyte differentiation by MMP-13 inhibitor small molecule encapsulated in alginate-chondroitin sulfate-platelet lysate hydrogel. Stem Cell Res Ther 2020; 11:436. [PMID: 33036643 PMCID: PMC7545577 DOI: 10.1186/s13287-020-01930-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 09/08/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells are a promising cell source for chondrogenic differentiation and have been widely used in several preclinical and clinical studies. However, they are prone to an unwanted differentiation process towards hypertrophy that limits their therapeutic efficacy. Matrix metallopeptidase 13 (MMP-13) is a well-known factor regulated during this undesirable event. MMP-13 is a collagen degrading enzyme, which is also highly expressed in the hypertrophic zone of the growth plate and in OA cartilage. Accordingly, we investigated the effect of MMP-13 inhibition on MSC hypertrophy. METHODS In this study, 5-bromoindole-2-carboxylic acid (BICA) was used as an inhibitory agent for MMP-13 expression. After identifying its optimal concentration, BICA was mixed into a hydrogel and the release rate was studied. To prepare the ideal hydrogel, chondroitin sulfate (CS) and platelet lysate (PL) were mixed with sodium alginate (Alg) at concentrations selected based on synergistic mechanical and rheometric properties. Then, four hydrogels were prepared by combining alginate (1.5%w/v) and/or CS (1%w/v) and/or PL (20%v/v). The chondrogenic potential and progression to hypertrophy of human bone marrow-derived mesenchymal stem cell (hBM-MSC)-loaded hydrogels were investigated under free swelling and mechanical loading conditions, in the presence and absence of BICA. RESULTS Viability of hBM-MSCs seeded in the four hydrogels was similar. qRT-PCR revealed that BICA could successfully inhibit MMP-13 expression, which led to an inhibition of Coll X and induction of Coll-II, in both free swelling and loading conditions. The GAG deposition was higher in the group combining BICA and mechanical stimulation. CONCLUSIONS It is concluded that BICA inhibition of MMP-13 reduces MSC hypertrophy during chondrogenesis.
Collapse
Affiliation(s)
- Shahrbanoo Jahangir
- Department of Tissue engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - David Eglin
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
| | - Naomi Pötter
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
- Department of orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center Albert-Ludwigs University, Albert-Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Mojtaba Khozaei Ravari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Martin J Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
- Department of orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center Albert-Ludwigs University, Albert-Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Ali Samadikuchaksaraei
- Department of Tissue engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mauro Alini
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland.
| | - Mohammadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Majid Safa
- Department of Tissue engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Hematology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Refaai MA, Conley GW, Hudson CA, Spinelli SL, Phipps RP, Morrell CN, Blumberg N, McRae HL. Evaluation of the procoagulant properties of a newly developed platelet modified lysate product. Transfusion 2020; 60:1579-1589. [PMID: 32415759 DOI: 10.1111/trf.15844] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Platelet transfusion is associated with logistical problems with the national storage guidelines of platelets. This results in decreased function in vivo as a result of the platelet storage lesion, and complications such as allergic or hemolytic reactions and thrombosis. We evaluated a new, freshly prepared platelet modified lysate (PML) product designed to be more procoagulant than fresh and stored platelets. METHODS Fresh platelets were concentrated, sonicated, and centrifuged to produce PML. Samples of both washed and unwashed PML were evaluated for particle size, concentration, and activity, and then tested for clot kinetics and thrombin generation. PML samples were also stored at various temperatures for durations up to 6 months and evaluated for clot kinetics and thrombin generation throughout. RESULTS PML showed significantly higher concentration of platelet microparticles, increased procoagulant properties, and increased thrombin generation as compared to fresh and stored platelets. In addition, PML maintained its clot kinetics over a 6-month storage period with variable storage conditions. CONCLUSIONS The newly proposed PML product is more procoagulant, stable, and has additional potential applications than currently available platelet products. Further studies will be performed to assess its functions in vivo and to assess thrombotic potential.
Collapse
Affiliation(s)
- Majed A Refaai
- Transfusion Medicine Division, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Grace W Conley
- Transfusion Medicine Division, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Sherry L Spinelli
- Transfusion Medicine Division, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Richard P Phipps
- Department of Environmental Medicine and Microbiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Craig N Morrell
- Department of Cardiovascular Research, Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Neil Blumberg
- Transfusion Medicine Division, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Hannah L McRae
- Transfusion Medicine Division, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
14
|
Faccendini A, Ruggeri M, Miele D, Rossi S, Bonferoni MC, Aguzzi C, Grisoli P, Viseras C, Vigani B, Sandri G, Ferrari F. Norfloxacin-Loaded Electrospun Scaffolds: Montmorillonite Nanocomposite vs. Free Drug. Pharmaceutics 2020; 12:pharmaceutics12040325. [PMID: 32260441 PMCID: PMC7238150 DOI: 10.3390/pharmaceutics12040325] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
Infections in nonhealing wounds remain one of the major challenges. Recently, nanomedicine approach seems a valid option to overcome the antibiotic resistance mechanisms. The aim of this study was the development of three types of polysaccharide-based scaffolds (chitosan-based (CH), chitosan/chondroitin sulfate-based (CH/CS), chitosan/hyaluronic acid-based (CH/HA)), as dermal substitutes, to be loaded with norfloxacin, intended for the treatment of infected wounds. The scaffolds have been loaded with norfloxacin as a free drug (N scaffolds) or in montmorillonite nanocomposite (H—hybrid-scaffolds). Chitosan/glycosaminoglycan (chondroitin sulfate or hyaluronic acid) scaffolds were prepared by means of electrospinning with a simple, one-step process. The scaffolds were characterized by 500 nm diameter fibers with homogeneous structures when norfloxacin was loaded as a free drug. On the contrary, the presence of nanocomposite caused a certain degree of surface roughness, with fibers having 1000 nm diameters. The presence of norfloxacin–montmorillonite nanocomposite (1%) caused higher deformability (90–120%) and lower elasticity (5–10 mN/cm2), decreasing the mechanical resistance of the systems. All the scaffolds were proven to be degraded via lysozyme (this should ensure scaffold resorption) and this sustained the drug release (from 50% to 100% in 3 days, depending on system composition), especially when the drug was loaded in the scaffolds as a nanocomposite. Moreover, the scaffolds were able to decrease the bioburden at least 100-fold, proving that drug loading in the scaffolds did not impair the antimicrobial activity of norfloxacin. Chondroitin sulfate and montmorillonite in the scaffolds are proven to possess a synergic performance, enhancing the fibroblast proliferation without impairing norfloxacin’s antimicrobial properties. The scaffold based on chondroitin sulfate, containing 1% norfloxacin in the nanocomposite, demonstrated adequate stiffness to sustain fibroblast proliferation and the capability to sustain antimicrobial properties to prevent/treat nonhealing wound infection during the healing process.
Collapse
Affiliation(s)
- Angela Faccendini
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (A.F.); (M.R.); (D.M.); (S.R.); (M.C.B.); (P.G.); (B.V.); (F.F.)
| | - Marco Ruggeri
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (A.F.); (M.R.); (D.M.); (S.R.); (M.C.B.); (P.G.); (B.V.); (F.F.)
| | - Dalila Miele
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (A.F.); (M.R.); (D.M.); (S.R.); (M.C.B.); (P.G.); (B.V.); (F.F.)
| | - Silvia Rossi
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (A.F.); (M.R.); (D.M.); (S.R.); (M.C.B.); (P.G.); (B.V.); (F.F.)
| | - Maria Cristina Bonferoni
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (A.F.); (M.R.); (D.M.); (S.R.); (M.C.B.); (P.G.); (B.V.); (F.F.)
| | - Carola Aguzzi
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Campus of Cartuja, 18071 Granada, Spain; (C.A.); (C.V.)
| | - Pietro Grisoli
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (A.F.); (M.R.); (D.M.); (S.R.); (M.C.B.); (P.G.); (B.V.); (F.F.)
| | - Cesar Viseras
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Campus of Cartuja, 18071 Granada, Spain; (C.A.); (C.V.)
| | - Barbara Vigani
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (A.F.); (M.R.); (D.M.); (S.R.); (M.C.B.); (P.G.); (B.V.); (F.F.)
| | - Giuseppina Sandri
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (A.F.); (M.R.); (D.M.); (S.R.); (M.C.B.); (P.G.); (B.V.); (F.F.)
- Correspondence: ; Tel.: +0039-0382-987728
| | - Franca Ferrari
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (A.F.); (M.R.); (D.M.); (S.R.); (M.C.B.); (P.G.); (B.V.); (F.F.)
| |
Collapse
|
15
|
Bonferoni MC, Rossi S, Sandri G, Caramella C, Del Fante C, Perotti C, Miele D, Vigani B, Ferrari F. Bioactive Medications for the Delivery of Platelet Derivatives to Skin Wounds. Curr Drug Deliv 2019; 16:472-483. [PMID: 30894109 PMCID: PMC6637103 DOI: 10.2174/1381612825666190320154406] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/24/2019] [Accepted: 03/13/2019] [Indexed: 12/13/2022]
Abstract
Chronic wounds are the result of alterations in the complex series of events of physiological wound healing. In particular, the prolonged inflammation results in increased protease activity, in the deg-radation of extracellular matrix (ECM) and of growth factors (GFs). The relevance of platelet GFs in maintaining and restoring the complex equilibrium of different moments in wound healing is well recog-nized. Moreover, the observed decrease of their levels in chronic wounds suggested a possible therapeutic role of the external application to the wounds. It has been also pointed out that tissue regeneration can be more efficiently obtained by the synergic use of different GFs. Platelet derivatives such as platelet-rich plasma (PRP) and platelet lysate (PL) are able to release GFs in a balanced pool. Their therapeutic use in regenerative medicine and wound healing has been therefore more and more frequently proposed in clini-cal trials and in the literature. The development of a suitable formulation able to control the GFs release rate, to protect the GFs, and to assure their prolonged contact with the wound site, is of paramount im-portance for the therapeutic success. The present review considers some formulation approaches for PRP and PL application to wounds
Collapse
Affiliation(s)
| | - Silvia Rossi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Giuseppina Sandri
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Carla Caramella
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Claudia Del Fante
- Immunohaematology and Transfusion Service and Cell Therapy Unit of Fondazione IRCCS, S. Matteo, 27100 Pavia, Italy
| | - Cesare Perotti
- Immunohaematology and Transfusion Service and Cell Therapy Unit of Fondazione IRCCS, S. Matteo, 27100 Pavia, Italy
| | - Dalila Miele
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Barbara Vigani
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Franca Ferrari
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
16
|
Burnouf T, Barro L, Nebie O, Wu YW, Goubran H, Knutson F, Seghatchian J. Viral safety of human platelet lysate for cell therapy and regenerative medicine: Moving forward, yes, but without forgetting the past. Transfus Apher Sci 2019; 58:102674. [PMID: 31735652 DOI: 10.1016/j.transci.2019.102674] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Growth factor-rich pooled human platelet lysate (HPL), made from human platelet concentrates, is one new blood-derived bioproduct that is attracting justified interest as a xeno-free supplement of growth media for human cell propagation for cell therapy. HPL can also find potentially relevant applications in the field of regenerative medicine. Therefore, the therapeutic applications of HPL go far beyond the standard clinical applications of the traditional blood products typically used in patients suffering from life-threatening congenital or acquired deficiencies in cellular components or proteins due to severe genetic diseases or trauma. A wider population of patients, suffering from various pathologies than has traditionally been the case, is thus, now susceptible to receiving a human blood-derived product. These patients would, therefore, be exposed to the possible, but avoidable, side effects of blood products, including transfusion-transmitted infections, most specifically virus transmissions. Unfortunately, not all manufacturers, suppliers, and users of HPL may have a strong background in the blood product industry. As such, they may not be fully aware of the various building blocks that should contribute to the viral safety of HPL as is already the case for any licensed blood products. The purpose of this manuscript is to reemphasize all the measures, including in regulatory aspects, capable of assuring that HPL exhibits a sufficient pathogen safety margin, especially when made from large pools of human platelet concentrates. It is vital to remember the past to avoid that the mistakes, which happened 30 to 40 years ago and led to the contamination of many blood recipients, be repeated due to negligence or ignorance of the facts.
Collapse
Affiliation(s)
- Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; Research Center of Biomedical Devices, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Lassina Barro
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Ouada Nebie
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wen Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Hadi Goubran
- Saskatoon Cancer Centre, Division of Oncology, College of Medicine, University of Saskatchewan, Canada
| | - Folke Knutson
- Clinical Immunology and Transfusion Medicine IGP, Uppsala University, Uppsala, Sweden
| | - Jerard Seghatchian
- International Consultancy in Blood Components Quality/Safety Improvement, Audit/Inspection and DDR Strategies, London, UK
| |
Collapse
|
17
|
Sandri G, Rossi S, Bonferoni MC, Miele D, Faccendini A, Del Favero E, Di Cola E, Icaro Cornaglia A, Boselli C, Luxbacher T, Malavasi L, Cantu’ L, Ferrari F. Chitosan/glycosaminoglycan scaffolds for skin reparation. Carbohydr Polym 2019; 220:219-227. [DOI: 10.1016/j.carbpol.2019.05.069] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/11/2019] [Accepted: 05/23/2019] [Indexed: 01/01/2023]
|
18
|
Costa-Almeida R, Calejo I, Altieri R, Domingues RMA, Giordano E, Reis RL, Gomes ME. Exploring platelet lysate hydrogel-coated suture threads as biofunctional composite living fibers for cell delivery in tissue repair. ACTA ACUST UNITED AC 2019; 14:034104. [PMID: 30844766 DOI: 10.1088/1748-605x/ab0de6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
To engineer functional tissue substitutes, it is required a multi-component, multi-scale approach that combines both physical, chemical and biological cues. Fiber-based techniques have been explored in the field of tissue engineering to produce structures recapitulating tissue architecture and mechanical properties. In this work, we engineered biofunctional composite living fibers (CLF) as multi-compartment fibers with a mechanically competent core and a hydrogel layer. For this purpose, commercial silk suture threads were coated with a platelet lysate (PL) hydrogel by first embedding the threads in a thrombin solution and then incubating in PL. The fabrication set-up was optimized and the biological performance was studied by encapsulating human adipose-derived stem cells (hASCs). The developed coating process rendered CLF with a homogenous PL hydrogel layer covering suture threads. Encapsulated hASCs were viable up to 14 d in culture and were able to align at the surface of the core fiber and deposit collagen types I and III. In summary, the study shows that PL-hASCs hydrogel coated suture threads represent a simple multi-compartment and multifunctional system, with PL hydrogel offering biofunctionality to guide the biological activities of encapsulated cells in addition to the replication of tissue-level mechanical support provided by the suture threads.
Collapse
Affiliation(s)
- Raquel Costa-Almeida
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | | | | | | | | | | |
Collapse
|
19
|
Saporito F, Baugh LM, Rossi S, Bonferoni MC, Perotti C, Sandri G, Black L, Ferrari F. In Situ Gelling Scaffolds Loaded with Platelet Growth Factors to Improve Cardiomyocyte Survival after Ischemia. ACS Biomater Sci Eng 2018; 5:329-338. [PMID: 33405861 DOI: 10.1021/acsbiomaterials.8b01064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Myocardial infarction is caused by prolonged ischemia and it is one of the main cause that leads to heart failures. The aim of the present work was the development of in situ gelling systems, based on poloxamer 407 (P407) or sodium alginate (Alg), loaded with platelet lysate (PL) to enhance cardiomyocyte survival after ischemia. Chondroitin sulfate (CS), a negatively charged glycosaminoglycan able to interact with different positively charged bioactive molecules, such as growth factors, was also investigated with both the systems. The gelation properties of both systems (viscosity, viscoelasticity, consistency by means of penetrometry, and injectability) were characterized in a physiological environment. In vitro evaluation of biocompatibility using fetal cardiac cells (cardiomyocytes and cardiac fibroblasts) demonstrated that the PL loaded alginate/chondroitin sulfate system retained the highest number of viable cells with equal distribution of the populations of cardiomyocytes and fibroblasts. Furthermore, the ability of the systems to improve cardiomyocyte survival after ischemia was also assessed. PL allowed for the highest degree of survival of cardiomyocytes after oxidative damage (simulating ischemic conditions due to MI) and both the Alg + CS PL and, to a greater extent, the PL alone demonstrated a considerable increase in survival of cardiomyocytes. In conclusion, an in situ gelling alginate-chondroitin sulfate system, loaded with platelet lysate, was able to improve the survival of cardiomyocytes after oxidative damage resulting in a promising system to improve cardiac cell viability after ischemia.
Collapse
Affiliation(s)
- Francesca Saporito
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Lauren M Baugh
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Silvia Rossi
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | | | - Cesare Perotti
- Immunohaematology and Transfusion Service, Apheresis and Cell Therapy Unit, Fondazione IRCCS Policlinico S. Matteo, Viale Golgi 19, Pavia 27100, Italy
| | - Giuseppina Sandri
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Lauren Black
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Franca Ferrari
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| |
Collapse
|
20
|
Rossi S, Mori M, Vigani B, Bonferoni MC, Sandri G, Riva F, Caramella C, Ferrari F. A novel dressing for the combined delivery of platelet lysate and vancomycin hydrochloride to chronic skin ulcers: Hyaluronic acid particles in alginate matrices. Eur J Pharm Sci 2018; 118:87-95. [PMID: 29574078 DOI: 10.1016/j.ejps.2018.03.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/26/2018] [Accepted: 03/20/2018] [Indexed: 11/19/2022]
Abstract
The aim of the present work was to develop a medication allowing for the combined delivery of platelet lysate (PL) and an anti-infective model drug, vancomycin hydrochloride (VCM), to chronic skin ulcers. A simple method was set up for the preparation of hyaluronic acid (HA) core-shell particles, loaded with PL and coated with calcium alginate, embedded in a VCM containing alginate matrix. Two different CaCl2 concentrations were investigated to allow for HA/PL core-shell particle formation. The resulting dressings were characterized for mechanical and hydration properties and tested in vitro (on fibroblasts) and ex-vivo (on skin biopsies) for biological activity. They were found of sufficient mechanical strength to withstand packaging and handling stress and able to absorb a high amount of wound exudate and to form a protective gel on the lesion area. The CaCl2 concentration used for shell formation did not affect VCM release from the alginate matrix, but strongly modified the release of PGFAB (chosen as representative of growth factors present in PL) from HA particles. In vitro and ex vivo tests provided sufficient proof of concept of the ability of dressings to improve skin ulcers healing.
Collapse
Affiliation(s)
- S Rossi
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - M Mori
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - B Vigani
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - M C Bonferoni
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - G Sandri
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - F Riva
- Department of Public Health, Experimental and Forensic Medicine, Histology and Embryology Unit, University of Pavia 10, 27100 Pavia, Italy
| | - C Caramella
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - F Ferrari
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| |
Collapse
|
21
|
Saporito F, Sandri G, Rossi S, Bonferoni MC, Riva F, Malavasi L, Caramella C, Ferrari F. Freeze dried chitosan acetate dressings with glycosaminoglycans and traxenamic acid. Carbohydr Polym 2018; 184:408-417. [DOI: 10.1016/j.carbpol.2017.12.066] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/17/2017] [Accepted: 12/24/2017] [Indexed: 01/01/2023]
|
22
|
Costa-Almeida R, Franco AR, Pesqueira T, Oliveira MB, Babo PS, Leonor IB, Mano JF, Reis RL, Gomes ME. The effects of platelet lysate patches on the activity of tendon-derived cells. Acta Biomater 2018; 68:29-40. [PMID: 29341933 DOI: 10.1016/j.actbio.2018.01.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/04/2017] [Accepted: 01/09/2018] [Indexed: 02/06/2023]
Abstract
Platelet-derived biomaterials are widely explored as cost-effective sources of therapeutic factors, holding a strong potential for endogenous regenerative medicine. Particularly for tendon repair, treatment approaches that shift the injury environment are explored to accelerate tendon regeneration. Herein, genipin-crosslinked platelet lysate (PL) patches are proposed for the delivery of human-derived therapeutic factors in patch augmentation strategies aiming at tendon repair. Developed PL patches exhibited a controlled release profile of PL proteins, including bFGF and PDGF-BB. Additionally, PL patches exhibited an antibacterial effect by preventing the adhesion, proliferation and biofilm formation by S. aureus, a common pathogen in orthopaedic surgical site infections. Furthermore, these patches supported the activity of human tendon-derived cells (hTDCs). Cells were able to proliferate over time and an up-regulation of tenogenic genes (SCX, COL1A1 and TNC) was observed, suggesting that PL patches may modify the behavior of hTDCs. Accordingly, hTDCs deposited tendon-related extracellular matrix proteins, namely collagen type I and tenascin C. In summary, PL patches can act as a reservoir of biomolecules derived from PL and support the activity of native tendon cells, being proposed as bioinstructive patches for tendon regeneration. STATEMENT OF SIGNIFICANCE Platelet-derived biomaterials hold great interest for the delivery of therapeutic factors for applications in endogenous regenerative medicine. In the particular case of tendon repair, patch augmentation strategies aiming at shifting the injury environment are explored to improve tendon regeneration. In this study, PL patches were developed with remarkable features, including the controlled release of growth factors and antibacterial efficacy. Remarkably, PL patches supported the activity of native tendon cells by up-regulating tenogenic genes and enabling the deposition of ECM proteins. This patch holds great potential towards simultaneously reducing post-implantation surgical site infections and promoting tendon regeneration for prospective in vivo applications.
Collapse
|
23
|
Saporito F, Sandri G, Bonferoni MC, Rossi S, Malavasi L, Fante CD, Vigani B, Black L, Ferrari F. Electrospun Gelatin⁻Chondroitin Sulfate Scaffolds Loaded with Platelet Lysate Promote Immature Cardiomyocyte Proliferation. Polymers (Basel) 2018; 10:polym10020208. [PMID: 30966244 PMCID: PMC6415061 DOI: 10.3390/polym10020208] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 02/15/2018] [Accepted: 02/20/2018] [Indexed: 12/11/2022] Open
Abstract
The aim of the present work was the development of heart patches based on gelatin (G) and chondroitin sulfate (CS) to be used as implants to improve heart recovery after corrective surgery for critical congenital heart defects (CHD). Patches were prepared by means of electrospinning to obtain nanofibrous scaffolds and they were loaded with platelet lysate (PL) as a source of growth factors to further enhance the repair process. Scaffolds were characterized for morphology and mechanical properties and for the capability to support in vitro adhesion and proliferation of dermal fibroblasts in order to assess the system’s general biocompatibility. Adhesion and proliferation of endothelial cells and cardiac cells (cardiomyocytes and cardiac fibroblasts from rat fetuses) onto PL-loaded patches was evaluated. Patches presented good elasticity and high stiffness suitable for in vivo adaptation to heart contraction. CS improved adhesion and proliferation of dermal fibroblasts, as proof of their biocompatibility. Moreover, they enhanced the adhesion and proliferation of endothelial cells, a crucial mediator of cardiac repair. Cell adhesion and proliferation could be related to elastic properties, which could favor cell motility. The presence of platelet lysate and CS was crucial for the adhesion and proliferation of cardiac cells and, in particular, of cardiomyocytes: G/CS scaffold embedded with PL appeared to selectively promote proliferation in cardiomyocytes but not cardiac fibroblasts. In conclusion, G/CS scaffold seems to be a promising system to assist myocardial-repair processes in young patient, preserving cardiomyocyte viability and preventing cardiac fibroblast proliferation, likely reducing subsequent uncontrolled collagen deposition by fibroblasts following repair.
Collapse
Affiliation(s)
- Francesca Saporito
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Giuseppina Sandri
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | | | - Silvia Rossi
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Lorenzo Malavasi
- Department of Chemistry, Physical Chemistry Section, University of Pavia, Viale Taramelli 16, 27100 Pavia, Italy.
| | - Claudia Del Fante
- Immunohaematology and Transfusion Service, Apheresis and Cell Therapy Unit, Fondazione IRCCS Policlinico S. Matteo, Viale Golgi 19, Pavia 27100, Italy.
| | - Barbara Vigani
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Lauren Black
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
| | - Franca Ferrari
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| |
Collapse
|
24
|
Comparative Analysis of Different Platelet Lysates and Platelet Rich Preparations to Stimulate Tendon Cell Biology: An In Vitro Study. Int J Mol Sci 2018; 19:ijms19010212. [PMID: 29320421 PMCID: PMC5796161 DOI: 10.3390/ijms19010212] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/05/2018] [Accepted: 01/08/2018] [Indexed: 02/07/2023] Open
Abstract
The poor healing potential of tendons is still a clinical problem, and the use of Platelet Rich Plasma (PRP) was hypothesized to stimulate healing. As the efficacy of PRPs remains unproven, platelet lysate (PL) could be an alternative with its main advantages of storage and characterization before use. Five different blood products were prepared from 16 male donors: human serum, two PRPs (Arthrex, (PRP-ACP); RegenLab (PRP-BCT)), platelet concentrate (apheresis, PC), and PL (freezing-thawing destruction of PC). Additionally, ten commercial allogenic PLs (AlloPL) from pooled donors were tested. The highest concentration of most growth factors was found in AlloPL, whereas the release of growth factors lasted longer in the other products. PRP-ACP, PRP-BCT, and PC significantly increased cell viability of human tenocyte-like cells, whereas PC and AlloPL increased Col1A1 expression and PRP-BCT increased Col3A1 expression. MMP-1, IL-1β, and HGF expression was significantly increased and Scleraxis expression decreased by most blood products. COX1 expression significantly decreased by PC and AlloPL. No clear positive effects on tendon cell biology could be shown, which might partially explain the weak outcome results in clinical practice. Pooled PL seemed to have the most beneficial effects and might be the future in using blood products for tendon tissue regeneration.
Collapse
|
25
|
Saporito F, Sandri G, Bonferoni MC, Rossi S, Boselli C, Icaro Cornaglia A, Mannucci B, Grisoli P, Vigani B, Ferrari F. Essential oil-loaded lipid nanoparticles for wound healing. Int J Nanomedicine 2017; 13:175-186. [PMID: 29343956 PMCID: PMC5747963 DOI: 10.2147/ijn.s152529] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Chronic wounds and severe burns are diseases responsible for severe morbidity and even death. Wound repair is a crucial process and tissue regeneration enhancement and infection prevention are key factors to minimize pain, discomfort, and scar formation. The aim of this work was the development of lipid nanoparticles (solid lipid nanoparticles and nanostructured lipid carriers [NLC]), to be loaded with eucalyptus or rosemary essential oils and to be used, as medical devices, to enhance healing of skin wounds. Lipid nanoparticles were based on natural lipids: cocoa butter, as solid lipid, and olive oil or sesame oil, as liquid lipids. Lecithin was chosen as surfactant to stabilize nanoparticles and to prevent their aggregation. The systems were prepared by high shear homogenization followed by ultrasound application. Nanoparticles were characterized for physical–chemical properties, bioadhesion, cytocompatibility, in vitro proliferation enhancement, and wound healing properties toward normal human dermal fibroblasts. Antimicrobial activity of nanoparticles was evaluated against two reference microbial strains, one of Staphylococcus aureus, the other of Streptococcus pyogenes. Finally, the capability of nanoparticles to promote wound healing in vivo was evaluated on a rat burn model. NLC based on olive oil and loaded with eucalyptus oil showed appropriate physical–chemical properties, good bioadhesion, cytocompatibility, in vitro proliferation enhancement, and wound healing properties toward fibroblasts, associated to antimicrobial properties. Moreover, the in vivo results evidenced the capability of these NLC to enhance the healing process. Olive oil, which is characterized by a high content of oleic acid, proved to exert a synergic effect with eucalyptus oil with respect to antimicrobial activity and wound repair promotion.
Collapse
Affiliation(s)
| | | | | | - Silvia Rossi
- Department of Drug Sciences, University of Pavia, Pavia
| | | | | | | | | | | | | |
Collapse
|
26
|
Menter DG, Kopetz S, Hawk E, Sood AK, Loree JM, Gresele P, Honn KV. Platelet "first responders" in wound response, cancer, and metastasis. Cancer Metastasis Rev 2017; 36:199-213. [PMID: 28730545 PMCID: PMC5709140 DOI: 10.1007/s10555-017-9682-0] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Platelets serve as "first responders" during normal wounding and homeostasis. Arising from bone marrow stem cell lineage megakaryocytes, anucleate platelets can influence inflammation and immune regulation. Biophysically, platelets are optimized due to size and discoid morphology to distribute near vessel walls, monitor vascular integrity, and initiate quick responses to vascular lesions. Adhesion receptors linked to a highly reactive filopodia-generating cytoskeleton maximizes their vascular surface contact allowing rapid response capabilities. Functionally, platelets normally initiate rapid clotting, vasoconstriction, inflammation, and wound biology that leads to sterilization, tissue repair, and resolution. Platelets also are among the first to sense, phagocytize, decorate, or react to pathogens in the circulation. These platelet first responder properties are commandeered during chronic inflammation, cancer progression, and metastasis. Leaky or inflammatory reaction blood vessel genesis during carcinogenesis provides opportunities for platelet invasion into tumors. Cancer is thought of as a non-healing or chronic wound that can be actively aided by platelet mitogenic properties to stimulate tumor growth. This growth ultimately outstrips circulatory support leads to angiogenesis and intravasation of tumor cells into the blood stream. Circulating tumor cells reengage additional platelets, which facilitates tumor cell adhesion, arrest and extravasation, and metastasis. This process, along with the hypercoagulable states associated with malignancy, is amplified by IL6 production in tumors that stimulate liver thrombopoietin production and elevates circulating platelet numbers by thrombopoiesis in the bone marrow. These complex interactions and the "first responder" role of platelets during diverse physiologic stresses provide a useful therapeutic target that deserves further exploration.
Collapse
Affiliation(s)
- David G Menter
- Department of Gastrointestinal Medical Oncology, M. D. Anderson Cancer Center, Room#: FC10.3004, 1515 Holcombe Boulevard--Unit 0426, Houston, TX, 77030, USA.
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, M. D. Anderson Cancer Center, Room#: FC10.3004, 1515 Holcombe Boulevard--Unit 0426, Houston, TX, 77030, USA
| | - Ernest Hawk
- Office of the Vice President Cancer Prevention & Population Science, M. D. Anderson Cancer Center, Unit 1370, 1515 Holcombe Boulevard, Houston, TX, 77054, USA
| | - Anil K Sood
- Gynocologic Oncology & Reproductive Medicine, M. D. Anderson Cancer Center, Unit 1362, 1515 Holcombe Boulevard, Houston, TX, 77054, USA
- Department of Cancer Biology, M. D. Anderson Cancer Center, Unit 1362, 1515 Holcombe Boulevard, Houston, TX, 77054, USA
- Center for RNA Interference and Non-Coding RNA The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Jonathan M Loree
- Department of Gastrointestinal Medical Oncology, M. D. Anderson Cancer Center, Room#: FC10.3004, 1515 Holcombe Boulevard--Unit 0426, Houston, TX, 77030, USA
| | - Paolo Gresele
- Department of Medicine, Section of Internal and Cardiovascular Medicine, University of Perugia, Via E. Dal Pozzo, 06126, Perugia, Italy
| | - Kenneth V Honn
- Bioactive Lipids Research Program, Department of Pathology, Wayne State University, 431 Chemistry Bldg, 5101 Cass Avenue, Detroit, MI, 48202, USA
- Department of Pathology, Wayne State University, 431 Chemistry Bldg, 5101 Cass Avenue, Detroit, MI, 48202, USA
- Cancer Biology Division, Wayne State University School of Medicine, 431 Chemistry Bldg, 5101 Cass Avenue, Detroit, MI, 48202, USA
| |
Collapse
|
27
|
Businaro R, Corsi M, Di Raimo T, Marasco S, Laskin DL, Salvati B, Capoano R, Ricci S, Siciliano C, Frati G, De Falco E. Multidisciplinary approaches to stimulate wound healing. Ann N Y Acad Sci 2016; 1378:137-142. [PMID: 27434638 DOI: 10.1111/nyas.13158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/01/2016] [Indexed: 01/08/2023]
Abstract
New civil wars and waves of terrorism are causing crucial social changes, with consequences in all fields, including health care. In particular, skin injuries are evolving as an epidemic issue. From a physiological standpoint, although wound repair takes place more rapidly in the skin than in other tissues, it is still a complex organ to reconstruct. Genetic and clinical variables, such as diabetes, smoking, and inflammatory/immunological pathologies, are also important risk factors limiting the regenerative potential of many therapeutic applications. Therefore, optimization of current clinical strategies is critical. Here, we summarize the current state of the field by focusing on stem cell therapy applications in wound healing, with an emphasis on current clinical approaches being developed. These involve protocols for the ex vivo expansion of adipose tissue-derived mesenchymal stem cells by means of a patented Good Manufacturing Practice-compliant platelet lysate. Combinations of multiple strategies, including genetic modifications and stem cells, biomimetic scaffolds, and novel vehicles, such as nanoparticles, are also discussed as future approaches.
Collapse
Affiliation(s)
- Rita Businaro
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.
| | - Mariangela Corsi
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Tania Di Raimo
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Sergio Marasco
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey
| | - Bruno Salvati
- Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
| | - Raffaele Capoano
- Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
| | - Serafino Ricci
- Department of Anatomical, Histological, Legal Medicine and Orthopedics Sciences, Sapienza University of Rome, Rome, Italy
| | - Camilla Siciliano
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Elena De Falco
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
28
|
Particulate systems based on pectin/chitosan association for the delivery of manuka honey components and platelet lysate in chronic skin ulcers. Int J Pharm 2016; 509:59-70. [DOI: 10.1016/j.ijpharm.2016.05.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/19/2016] [Accepted: 05/20/2016] [Indexed: 11/23/2022]
|
29
|
Platelet lysate and chondroitin sulfate loaded contact lenses to heal corneal lesions. Int J Pharm 2016; 509:188-196. [PMID: 27234702 DOI: 10.1016/j.ijpharm.2016.05.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 05/19/2016] [Accepted: 05/23/2016] [Indexed: 11/20/2022]
Abstract
Hemoderivative tear substitutes contain various ephiteliotrophic factors, such as growth factors (GF), involved in ocular surface homeostasis without immunogenic properties. The aim of the present work was the loading of platelet lysate into contact lenses to improve the precorneal permanence of platelet lysate growth factors on the ocular surface to enhance the treatment of corneal lesions. To this purpose, chondroitin sulfate, a sulfated glycosaminoglycan, which is normally present in the extracellular matrix, was associated with platelet lysate. In fact, chondroitin sulfate is capable of electrostatic interaction with positively charged growth factors, in particular, with bFGF, IGF, VEGF, PDGF and TGF-β, resulting in their stabilization and reduced degradation in solution. In the present work, various types of commercially available contact lenses have been loaded with chondroitin sulfate or chondroitin sulfate in association with platelet lysate to achieve a release of growth factors directly onto the corneal surface lesions. One type of contact lenses (PureVision(®)) showed in vitro good proliferation properties towards corneal cells and were able to enhance cut closure in cornea constructs.
Collapse
|
30
|
Collagen and Fractionated Platelet-Rich Plasma Scaffold for Dermal Regeneration. Plast Reconstr Surg 2016; 137:1498-1506. [DOI: 10.1097/prs.0000000000002094] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
31
|
|
32
|
Mori M, Rossi S, Ferrari F, Bonferoni MC, Sandri G, Riva F, Tenci M, Del Fante C, Nicoletti G, Caramella C. Sponge-Like Dressings Based on the Association of Chitosan and Sericin for the Treatment of Chronic Skin Ulcers. II. Loading of the Hemoderivative Platelet Lysate. J Pharm Sci 2016; 105:1188-95. [DOI: 10.1016/j.xphs.2015.11.043] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/21/2015] [Accepted: 11/23/2015] [Indexed: 01/25/2023]
|