Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 159
1.
Vet Med Sci ; 10(3): e1439, 2024 05.
Article En | MEDLINE | ID: mdl-38695208

This study evaluated the effect of ozone, chitosan-hyaluronic (Cs-HA) acid and mesenchymal stem cells (MSCs) on wound healing in rats. A total of 64 rats were randomly divided into four groups: control, ozone, Cs-HA + ozone and Cs-HA + ozone + MSCs. A 5 mm full-thickness wound was created on the back of each rat. The wound area was measured macroscopically on days 3, 5, 9 and 14. Tissue sections were prepared for histopathological evaluation of inflammation, collagen arrangement, neovascularization and epithelial tissue rearrangement. Macroscopic assessment showed differences in wound area on days 5, 9 and 14. Histopathological examination showed that the Cs-HA + ozone + MSCs and Cs-HA + ozone groups had significantly higher vascularization on day 3 compared to the ozone-treated and control groups. All treatment groups had significantly better collagen arrangement than the control group. On day 5, no significant difference was observed between different groups. On day 9, the inflammation level in the Cs-HA + ozone + MSCs group was significantly lower than in the other groups. All treatment groups had significantly better vascularization compared to the control group. On day 14, the rate of inflammation was significantly lower in the treatment groups than in the control group. Significantly higher collagen arrangement levels were observed in the Cs-HA + ozone and Cs-HA + ozone + MSCs groups compared to the control and ozone groups. All treatment groups had significantly better epithelial tissue rearrangement than the control group. Overall, the results of this study indicated that treatment with ozone, Cs-HA acid, Cs-HA and MSCs accelerated wound healing in rats. The effect of using Cs-HA acid with mesenchymal cells was better than the other types of treatment. Larger clinical trials are needed to assess these factors for improving chronic wound treatment.


Chitosan , Hyaluronic Acid , Mesenchymal Stem Cell Transplantation , Ozone , Wound Healing , Animals , Wound Healing/drug effects , Ozone/pharmacology , Rats , Hyaluronic Acid/pharmacology , Male , Mesenchymal Stem Cell Transplantation/veterinary , Rats, Wistar , Random Allocation
2.
J Am Vet Med Assoc ; 262(S1): S16-S23, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38565136

Despite a pressing need for new therapies to address unmet veterinary medical need, no approved stem cell products are available for use in cats in the US. To evaluate the current state of mesenchymal stem or stromal cell (MSC) research in cats, a scoping review of published literature was performed, which identified 108 publications related to feline MSCs. Twenty-six of the articles described administration of MSC products to a total of 215 cats. Twelve of the studies included a control group. These experimental and clinical trials used 7 cell sources, 9 administration routes, 12 delivery vehicles, and a 300-fold range in dosages for initial studies in healthy cats and cats with 12 naturally occurring and induced diseases. The majority of studies administered 2 doses of allogeneic, adipose-derived MSC IV and monitored a median of 6.5 treated cats for a median of 90 days. The majority (150/215 [69.8%]) of cats had no reported adverse events associated with treatment. Although an increase in feline MSC publications in the past 10 years indicates progress, the wide variety and small number of studies using MSCs and MSC products in cats demonstrates that current evaluations are mostly still in the discovery phase, and several issues remain related to larger scale trials using MSC products in cats. The current available publications provide information to direct further clinical study development and informed owner consent for study enrollment.


Cat Diseases , Mesenchymal Stem Cell Transplantation , Cats , Animals , Mesenchymal Stem Cell Transplantation/veterinary , Cat Diseases/therapy , Mesenchymal Stem Cells
3.
J Am Vet Med Assoc ; 262(S1): S24-S30, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38565137

A scoping review of published literature found 108 articles related to mesenchymal stem or stromal cell (MSC) use in cats. Twenty-four of the publications summarized the treatment of 192 cats with MSC products for 12 naturally occurring and induced diseases. These trials used a variety of cell sources, administration routes, delivery vehicles, and dosages. The majority of studies did not have a control group. The disease with the largest number of cats administered MSCs thus far is chronic kidney disease (n = 59 cats). The majority of cats had no adverse events associated with treatment, which supports continued interest in the potential use of MSC products to address unmet medical needs. Treatment outcomes of the 192 cats have ranged from no response to long-term cure, depending on the disease being treated and the particular study. Some of these early studies show promise and provide significant information to direct both the design and focus of larger clinical trials investigating the safety and efficacy of MSC treatment for veterinary and human applications.


Cat Diseases , Mesenchymal Stem Cell Transplantation , Cats , Animals , Cat Diseases/therapy , Mesenchymal Stem Cell Transplantation/veterinary , Mesenchymal Stem Cells
4.
BMC Vet Res ; 20(1): 110, 2024 Mar 18.
Article En | MEDLINE | ID: mdl-38500105

BACKGROUND: Traumatic brain injury (TBI) is a common condition in veterinary medicine that is difficult to manage.Veterinary regenerative therapy based on adipose mesenchymal stem cells seem to be an effective strategy for the treatment of traumatic brain injury. In this study, we evaluated therapeutic efficacy of canine Adipose-derived mesenchymal stem cells (AD-MSCs)in a rat TBI model, in terms of improved nerve function and anti-neuroinflammation. RESULTS: Canine AD-MSCs promoted neural functional recovery, reduced neuronal apoptosis, and inhibited the activation of microglia and astrocytes in TBI rats. According to the results in vivo, we further investigated the regulatory mechanism of AD-MSCs on activated microglia by co-culture in vitro. Finally, we found that canine AD-MSCs promoted their polarization to the M2 phenotype, and inhibited their polarization to the M1 phenotype. What's more, AD-MSCs could reduce the migration, proliferation and Inflammatory cytokines of activated microglia, which is able to inhibit inflammation in the central system. CONCLUSIONS: Collectively, the present study demonstrates that transplantation of canine AD-MSCs can promote functional recovery in TBI rats via inhibition of neuronal apoptosis, glial cell activation and central system inflammation, thus providing a theoretical basis for canine AD-MSCs therapy for TBI in veterinary clinic.


Brain Injuries, Traumatic , Dog Diseases , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Rodent Diseases , Rats , Animals , Dogs , Brain Injuries, Traumatic/therapy , Brain Injuries, Traumatic/veterinary , Microglia , Macrophages , Inflammation/veterinary , Mesenchymal Stem Cell Transplantation/veterinary , Mesenchymal Stem Cell Transplantation/methods
5.
J Am Vet Med Assoc ; 262(S1): S50-S60, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38471305

BACKGROUND: Platelet-rich plasma (PRP) and mesenchymal stromal or stem cells (MSCs) have been investigated as treatments for equine tendon and ligament injuries, but little consensus exists on the efficacy of these treatments. The study sought to evaluate the efficacy of PRP and MSC treatments by systematic review and meta-analysis. METHODS: A systematic review was performed using the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analysis) guidelines. Inclusion criteria required an original, peer-reviewed study where horses were administered MSCs or PRP (or both), and a comparator group was described. Studies were assessed for risk of bias and study quality. Random effects meta-analysis with inverse variance weighting was used to calculate pooled estimates of the ORs for the primary outcomes of return to performance and reinjury. RESULTS: The search criteria identified 764 unique studies, and 21 studies met the inclusion criteria for the systematic review. Seventeen studies were further assessed for the primary outcomes of return to performance and reinjury rate within a meta-analysis. Meta-analyses revealed no increase in the likelihood of a return to performance with any of the biologic treatments. However, MSCs and MSCs administered concurrently with PRP provide a reduced risk of reinjury. CLINICAL RELEVANCE: The current study identified a decrease in reinjury rate in horses administered MSCs or a combination of MSCs and PRP for tendinopathy and desmopathy. However, results should be interpreted with consideration of the heterogeneity of findings, poor study quality, and high risk of bias in the majority of studies.


Horse Diseases , Ligaments , Platelet-Rich Plasma , Tendon Injuries , Animals , Horses/injuries , Tendon Injuries/veterinary , Tendon Injuries/therapy , Ligaments/injuries , Horse Diseases/therapy , Mesenchymal Stem Cell Transplantation/veterinary
6.
J Am Vet Med Assoc ; 262(S1): S131-S140, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38479104

The discovery that a population of cells derived from adult tissues had regenerative properties and could be expanded and utilized therapeutically led to an abundance of research into the safety and efficacy of these cells for a variety of disease processes. Derived from tissues of mesenchymal origin, these cells were called mesenchymal stem cells or mesenchymal stromal cells (MSCs). MSCs were subsequently found to have a variety of anti-inflammatory, antimicrobial, and regenerative properties both in vitro and in vivo. These cells have been extensively studied and demonstrated to be effective in the treatment of autoimmune diseases, inflammatory diseases, infectious diseases, and other pathologic conditions. Multiple veterinary clinical trials have been performed for diseases such as osteoarthritis, soft tissue, and tendon injuries in addition to other inflammatory and infectious conditions. Exotic animal species can pose additional challenges to the treatment of common medical conditions such as osteoarthritis due to varying metabolism and tolerance to medications as well as potential difficulty to medicate or utilize other common forms of therapy such as physical therapy, laser, massage, and acupuncture. Current knowledge of MSC use in exotic species is limited, but there is a large potential for benefit to a variety of species utilizing this novel therapeutic tool. This review includes a brief overview of mechanisms and known applications of this therapy as well as current and potential applications of MSCs in exotic animal species.


Mesenchymal Stem Cell Transplantation , Animals , Mesenchymal Stem Cell Transplantation/veterinary , Animals, Exotic , Mesenchymal Stem Cells/cytology
7.
Vet J ; 304: 106090, 2024 04.
Article En | MEDLINE | ID: mdl-38417670

Inflammatory bowel disease (IBD) is a major subtype of chronic enteropathies in dogs and cats. Conventional drugs such as immunomodulatory medicines as glucocorticoids and/or other anti-inflammatory are mainly applied for treatment. However, these drugs are not always effective to maintain remission from IBD and are limited by unacceptable side effects. Hence, more effective and safe therapeutic options need to be developed. Mesenchymal stem cells (MSCs) are multipotent stem cells with a self-renewal capacity, and have immunomodulatory, anti-inflammatory, anti-fibrotic, and tissue repair properties. Therefore, the application of MSCs as an alternative therapy for IBD has great potential in veterinary medicine. The efficacy of adipose tissue-derived MSC (ADSC) therapy for IBD in dogs and cats has been reported, including numerous studies in animal models. However, treatment outcomes in clinical trials of human IBD patients have not been consistent with preclinical studies. MSC-based therapy for various diseases has received widespread attention, but various problems in such therapy remain, among which no consensus has been reached on the preparation and treatment procedures for MSCs, and cellular heterogeneity of MSCs may be an issue. This review describes the current status of ADSC therapy for canine and feline IBD and summarizes the cellular heterogeneity of canine ADSCs, to highlight the necessity for further reduction or elimination of MSCs heterogeneity and standardization of MSC-based therapies.


Cat Diseases , Dog Diseases , Inflammatory Bowel Diseases , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Cats , Animals , Dogs , Cat Diseases/therapy , Dog Diseases/therapy , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/veterinary , Mesenchymal Stem Cell Transplantation/veterinary , Mesenchymal Stem Cell Transplantation/methods , Anti-Inflammatory Agents
9.
J Am Vet Med Assoc ; 262(S1): S73-S82, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38295517

OBJECTIVE: To investigate mechanistically the reported beneficial effects of immune-activated mesenchymal stromal cell (MSC) therapy to treat equine septic arthritis, leveraging Nanostring technology. ANIMALS: 8 Quarter Horses with induced tibiotarsal Staphylococcus aureus septic arthritis treated IA with either Toll-like receptor-3 agonist polyinosinic:polycytidylic acid-activated MSCs + vancomycin antimicrobials (TLR-MSC-VAN; n = 4) or antimicrobials (VAN; 4). METHODS: Synovial tissues were collected and fixed in neutral-buffered 10% formalin, and formalin-fixed paraffin-embedded synovial and osteochondral tissues were sequenced using a custom-designed 200-gene equine Nanostring nCounter immune panel to directly quantify expression of key immune and cartilage-related genes. Immunohistochemistry to detect CD3+ T cells was performed on synovial tissues to further quantify T-cell infiltration in TLR-MSC-VAN- versus VAN-treated joints. RESULTS: Comparison of synovial transcriptomes between groups revealed moderate changes in differential gene expression, with upregulated expression of 9 genes and downregulated expression of 17 genes with fold change ≥ 2 or ≤ -2 and a significant false discovery rate-adjusted P value of ≤ .05. The most upregulated genes in TLR-MSC-VAN-treated horses included those related to T-lymphocyte recruitment and function, while pathways related to innate immune activation and inflammation were significantly downregulated. Immunohistochemistry and quantitation of CD3+ T-cell infiltrates revealed a numerically greater infiltrate in synovial tissues of TLR-MSC-VAN-treated horses, which did not reach statistical significance in this small sample set (P = .20). CLINICAL RELEVANCE: Targeted transcriptomic analyses using an equine Nanostring immune and cartilage health panel provided new mechanistic insights into how innate and adaptive immune cells within synovial tissues respond to TLR-activated MSC treatment when used to treat septic arthritis.


Arthritis, Infectious , Horse Diseases , Synovial Membrane , T-Lymphocytes , Animals , Horses , Arthritis, Infectious/veterinary , Horse Diseases/therapy , Horse Diseases/immunology , Synovial Membrane/cytology , Mesenchymal Stem Cells , Transcriptome , Staphylococcal Infections/veterinary , Gene Expression Profiling/veterinary , Female , Male , Mesenchymal Stem Cell Transplantation/veterinary
10.
Cells ; 12(21)2023 10 24.
Article En | MEDLINE | ID: mdl-37947591

The treatment of tendinopathies with multipotent mesenchymal stromal cells (MSCs) is a promising option in equine and human medicine. However, conclusive clinical evidence is lacking. The purpose of this study was to gain insight into clinical treatment efficacy and to identify suitable outcome measures for larger clinical studies. Fifteen horses with early naturally occurring tendon disease were assigned to intralesional treatment with allogeneic adipose-derived MSCs suspended in serum or with serum alone through block randomization (dosage adapted to lesion size). Clinicians and horse owners remained blinded to the treatment during 12 months (seven horses per group) and 18 months (seven MSC-group and five control-group horses) of follow-up including clinical examinations and diagnostic imaging. Clinical inflammation, lameness, and ultrasonography scores improved more over time in the MSC group. The lameness score difference significantly improved in the MSC group compared with the control group after 6 months. In the MSC group, five out of the seven horses were free of re-injuries and back to training until 12 and 18 months. In the control group, three out of the seven horses were free of re-injuries until 12 months. These results suggest that MSCs are effective for the treatment of early-phase tendon disease and provide a basis for a larger controlled study.


Hematopoietic Stem Cell Transplantation , Horse Diseases , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Reinjuries , Humans , Horses , Animals , Pilot Projects , Lameness, Animal/therapy , Lameness, Animal/pathology , Horse Diseases/therapy , Horse Diseases/pathology , Mesenchymal Stem Cell Transplantation/veterinary , Mesenchymal Stem Cells/pathology , Tendons/pathology
11.
Am J Vet Res ; 84(10): 1-8, 2023 Sep 01.
Article En | MEDLINE | ID: mdl-37669745

Tendon injuries are common in both veterinary and human clinical patients and result in morbidity, pain, and lost athletic performance. Consequently, utilizing naturally occurring injuries in veterinary patients as a comparative model could inform the development of novel therapies and increase translation for the treatment of human tendon injuries. Mesenchymal stem cells (MSCs) have shown considerable efficacy for the treatment of experimental and clinical superficial digital flexor tendon injury in the horse; however, the reinjury rate following treatment can remain high and MSC efficacy in treating other tendons is less well known. Additionally, the translation of MSC therapy to human tendon injury has remained poor. Recent evidence indicates that naïve MSC function can be enhanced through exogenous stimulation or manipulation of their environment. This stimulation or activation, herein termed MSC licensing, markedly alters MSC functions associated with immunomodulation, extracellular matrix remodeling, vascular development, bioactive factor production, and endogenous stromal/progenitor cell support. Additionally, a variety of licensing strategies has proven to influence MSC-secreted factors that have positively influenced outcome parameters in both in vitro and in vivo disease models separate from musculoskeletal tissues. Therefore, identifying the optimal licensing strategy for MSCs could ultimately provide an avenue for reliable and repeatable treatment of a broad range of tendon injuries of both veterinary and human clinical patients. This article details current evidence on the effects of licensed MSCs in both in vitro and in vivo disease models of different species and provides commentary on how those effector functions identified may be translated to the treatment of tendon injuries.


Horse Diseases , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Tendon Injuries , Humans , Animals , Horses , Mesenchymal Stem Cell Transplantation/veterinary , Tendon Injuries/therapy , Tendon Injuries/veterinary , Mesenchymal Stem Cells/physiology , Tendons , Extracellular Matrix
12.
Vet Clin North Am Equine Pract ; 39(3): 475-487, 2023 Dec.
Article En | MEDLINE | ID: mdl-37625917

Mesenchymal stem cells (MSCs) are used as a regenerative therapy in horses for musculoskeletal injury since the late 1990s and in some regions are standard of care for certain injuries. Yet, there is no Food and Drug Administration-approved MSC therapeutic in the United States for horses. In humans, lack of regulatory approval in the United States has been caused by failure of late-phase clinical trials to demonstrate consistent efficacy, perhaps because of nonuniformity of MSC preparation and application techniques. This article discusses clinical evidence for musculoskeletal applications of MSCs in the horse and current challenges to marketing approval.


Horse Diseases , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Musculoskeletal Diseases , Humans , Horses , Animals , Mesenchymal Stem Cell Transplantation/veterinary , Mesenchymal Stem Cell Transplantation/methods , Horse Diseases/therapy , Musculoskeletal Diseases/therapy , Musculoskeletal Diseases/veterinary
13.
J Vet Sci ; 24(4): e52, 2023 Jul.
Article En | MEDLINE | ID: mdl-37532297

BACKGROUND: Mesenchymal stem cells (MSCs) have been investigated as therapeutic agents for inflammatory bowel disease (IBD). Stimulation of MSCs with pro-inflammatory cytokines is an approach to enhance their immunomodulatory effects. However, further investigation is required to support their application in immune-mediated disorders and companion animals. OBJECTIVES: This study aimed to assess the therapeutic effect of tumor necrosis factor (TNF)-α-stimulated feline adipose tissue-derived MSCs (fAT-MSCs) in a dextran sulfate sodium (DSS)-induced colitis mouse model. METHODS: Colitis mice was made by drinking water with 3% DSS and fAT-MSCs were injected intraperitoneally. Colons were collected on day 10. The severity of the disease was evaluated and compared. Raw 264.7 cells were cultured with the conditioned medium to determine the mechanism, using quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assay. RESULTS: TNF-α-stimulated fAT-MSCs more improved severity of DSS-induced colitis in disease activity, colon length, histologic score, and inflammatory cytokine. In sectionized colon tissues, the group comprising TNF-α-stimulated fAT-MSCs had higher proportion of CD11b+CD206+ macrophages than in the other groups. In vitro, TNF-α-stimulation increased cyclooxygenase-2 (COX-2) expression and prostaglandin E2 (PGE2) secretion from fAT-MSCs. The conditioned medium from TNF-α-stimulated fAT-MSCs enhanced the expression of interleukin-10 and arginase-1 in LPS-activated Raw 264.7 cells. CONCLUSIONS: These results represent that TNF-α-stimulated fat-mscs ameliorate the inflamed colon more effectively. Furthermore, we demonstrated that the effectiveness was interlinked with the COX-2/PGE2 pathway.


Cat Diseases , Colitis , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Cats , Mice , Adipose Tissue , Cat Diseases/metabolism , Colitis/chemically induced , Colitis/therapy , Colitis/metabolism , Colitis/veterinary , Culture Media, Conditioned/adverse effects , Cyclooxygenase 2/genetics , Cytokines/metabolism , Dextran Sulfate/toxicity , Dinoprostone/metabolism , Disease Models, Animal , Mesenchymal Stem Cell Transplantation/veterinary , Mesenchymal Stem Cells/physiology , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/metabolism
14.
Vet Res Commun ; 47(4): 2055-2070, 2023 Dec.
Article En | MEDLINE | ID: mdl-37421548

Mesenchymal stem cells (MSCs) are a promising tool for treating immune disorders. However, the immunomodulatory effects of canine MSCs compared with other commercialized biologics for treating immune disorders have not been well studied. In this study we investigated the characteristics and immunomodulatory effects of canine amnion membrane (cAM)-MSCs. We examined gene expression of immune modulation and T lymphocytes from activated canine peripheral blood mononuclear cell (PBMC) proliferation. As a result, we confirmed that cAM-MSCs upregulated immune modulation genes (TGF-ß1, IDO1 and PTGES2) and suppressed the proliferation capacity of T cells. Moreover, we confirmed the therapeutic effect of cAM-MSCs compared with oclacitinib (OCL), the most commonly used Janus kinase (JAK) inhibitor, as a treatment for canine atopic dermatitis (AD) using a mouse AD model. As a result, we confirmed that cAM-MSCs with PBS treatment groups (passage 4, 6 and 8) compared with PBS only (PBS) though scores of dermatologic signs, tissue pathologic changes and inflammatory cytokines were significantly reduced. In particular, cAM-MSCs were more effective than OCL in the recovery of wound dysfunction, regulation of mast cell activity and expression level of immune modulation protein. Interestingly, subcutaneous injection of cAM-MSCs induced weight recovery, but oral administration of oclacitinib induced weight loss as a side effect. In conclusion, this study suggests that cAM-MSCs can be developed as a safe canine treatment for atopic dermatitis without side effects through effective regeneration and immunomodulation.


Dermatitis, Atopic , Dog Diseases , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Dogs , Dermatitis, Atopic/therapy , Dermatitis, Atopic/veterinary , Dermatitis, Atopic/metabolism , Amnion/metabolism , Leukocytes, Mononuclear , Mesenchymal Stem Cell Transplantation/veterinary , Immunomodulation , Regeneration , Dog Diseases/therapy
15.
Res Vet Sci ; 162: 104948, 2023 Sep.
Article En | MEDLINE | ID: mdl-37478792

Mesenchymal stem cells are safe and effective for treating joint injuries. However, the most suitable cell source remains controversial. This randomized controlled, double-blind study aimed to evaluate the potentials of rabbit allogeneic bone marrow- (BMSCs), adipose- (ASCs) and synovial membrane- (SDSCs) derived stem cells encapsulated in fibrin glue (FG) in vivo. The therapeutic properties of fibrin glue in critical-sized osteochondral defects (ODs) were also investigated. A 3 × 3 mm-sized OD was created in the femoral patellar groove on both knees of New Zealand rabbits, except from the left knees of the control group in which the OD was 2 × 3mm. The rabbits were randomly divided into four groups (right/left knee): 3 × 3 mm / 2 × 3 mm-sized OD control group, FG/FG with ASCs group, FG/FG with BMSCs group, FG/FG with SDSCs group. The International Cartilage Repair Society (ICRS) and the O'Driscoll scales were used to evaluate tissue characteristics after 12 weeks. FG promoted the production of reparative tissue with superior macroscopic features. Allogeneic MSCs combined with FG improved the macroscopic and histological scores more than the FG groups. The tissue in the SDSCs group was macroscopically and histologically better than the ASCs and BMSCs groups. The ICRS score differed among the SDSCs and the ASCs groups, while the empty critical-sized ODs were filled with inferior tissue compared to smaller ones. The preclinical feasibility of stem cells for OD regeneration in rabbits and the osteochondrogenic superiority of SDSCs was demonstrated. Additional tests and extended studies are required to reassure the long-term safety of these findings.


Cartilage, Articular , Hematopoietic Stem Cell Transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Rabbits , Animals , Cartilage, Articular/pathology , Fibrin Tissue Adhesive , Mesenchymal Stem Cell Transplantation/veterinary , Hematopoietic Stem Cell Transplantation/veterinary
16.
Vet Q ; 42(1): 224-230, 2022 Dec.
Article En | MEDLINE | ID: mdl-36336651

Osteoarthritis is a progressive degenerative disease affecting joints. It is associated with structural and functional changes that cause lameness and pain in dogs. Mesenchymal stem cells (MSCs) are considered an ideal therapeutic candidate for treating inflammatory musculoskeletal conditions due to their paracrine and immunomodulatory characteristics. They are delivered intravenously or as intra-articular injections for treating canine osteoarthritis. However, ex vivo studies have confirmed that the osteoarthritic synovial fluid is cytotoxic to cultured MSCs. Therefore, intra-articular transplantation of viable MSCs should be considered counterproductive since it minimizes cellular viability. Similarly, the intravenous administration of MSCs limits the therapeutic effects on the organ of interest since most of the administered cells get trapped in the lungs. Therefore, cell-free therapeutic strategies such as conditioned media and extracellular vesicles (EVs) can potentially become the future of MSC-based therapy in managing canine osteoarthritis. It overcomes the limitations of MSC-based therapy, such as tumor differentiation, immunogenicity, and pulmonary embolization, and has advantages like low immunogenicity and off-shelf availability. In addition, they eliminate problems such as low cell survival, transmission of infections, and unpredictable behavior of the transplanted MSCs, thereby acting as a safe alternative to cell-based therapeutics. However, very limited data is available on the efficacy and safety of cell-free therapy using MSCs for managing canine osteoarthritis. Therefore, large-scale, multicentric, randomized clinical controlled trials are required to establish the therapeutic efficacy and safety of MSC-based cell-free therapy in clinical cases of canine osteoarthritis.


Dog Diseases , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Osteoarthritis , Dogs , Animals , Mesenchymal Stem Cell Transplantation/veterinary , Osteoarthritis/therapy , Osteoarthritis/veterinary , Mesenchymal Stem Cells/pathology , Injections, Intra-Articular/veterinary , Pain/veterinary , Dog Diseases/therapy
17.
Am J Vet Res ; 83(10)2022 Aug 18.
Article En | MEDLINE | ID: mdl-35973004

OBJECTIVE: Mesenchymal stromal (stem) cells (MSCs) have been studied to treat many common orthopedic injuries in horses. However, there is limited information available on when and how to use this treatment effectively. The aim of this retrospective study is to report case features, treatment protocols, and clinical outcomes in horses treated with MSCs. ANIMALS: 65 horses presenting with tendinous, ligamentous, and articular injuries, and treated with MSCs prepared by a single laboratory between 2016 and 2019. Outcome information was available for 26 horses. PROCEDURES: Signalment, clinical signs, diagnostic methods, treatment protocol features (prior and concurrent therapies, cell origin, dose, application site and number), and effective outcomes were analyzed. The analysis was focused on comparing the effect of different MSC treatment protocols (eg, autologous vs allogeneic) on outcome rather than the effectiveness of MSC treatment. RESULTS: MSC treatment resulted in 59.1% (clinical lameness) to 76.9% (imaging structure) improvement in horses with diverse ages, breeds, sex, and lesions. The use of other therapeutic methods before MSC application (eg, anti-inflammatories, shockwave, laser, icing, resting, bandage and stack wrap, intra-articular injections, and/or surgical debridement) was shown to be statistically more effective compared to MSCs used as the primary therapeutic procedure (P < .05). Autologous versus allogeneic treatment outcomes were not significantly different. CLINICAL RELEVANCE: A prospective MSC treatment study with standardization and controls to evaluate the different features of MSC treatment protocols is needed. The various case presentations and treatment protocols evaluated can be used to inform practitioners who are currently using MSCs in clinical practice.


Horses/injuries , Mesenchymal Stem Cell Transplantation/veterinary , Mesenchymal Stem Cells/physiology , Animals , Injections, Intra-Articular/veterinary , Joints/injuries , Ligaments/injuries , Retrospective Studies , Tendon Injuries/therapy , Tendon Injuries/veterinary
18.
Vet Med Sci ; 8(5): 2086-2091, 2022 09.
Article En | MEDLINE | ID: mdl-35838746

BACKGROUND: Hepatocellular carcinoma (HCC) is the fifth most diagnosed cancer and the second leading cause of cancer-related deaths worldwide. Sorafenib is the standard treatment used in the advanced stages of HCC. Cell therapy with mesenchymal stem cells (MSCs)-based cell therapy has proven effective in immune regulation and tumour growth inhibition. OBJECTIVES: In this study, we investigated the anti-inflammatory effect of MSCs on HCC xenografts. METHODS: Human HepG2 cell lines were subcutaneously implanted into the flank of 12 nude mice, divided into three groups: the control group, the IV group (intravenous MSCs injection) and the local group (local MSCs injection). Mice were sacrificed 6 weeks after tumour implantation, and tumours were resected entirety. Quantitative real-time polymerase chain reaction (qRT-PCR) measured the gene expression of inflammatory markers, including tumour necrosis factor-α (TNF-α), interleukin (IL)-1α and IL-10. Aspartate transaminase (AST), alanine transaminase (ALT) and urea levels were measured using spectrophotometry to ensure the safety of MSC therapy. RESULTS: Gene expressions for all three inflammatory markers were reduced in both MSCs groups compared to the control group. AST, ALT and urea levels remained in normal ranges. CONCLUSIONS: MSC therapy can reduce inflammation in HCC xenograft mouse models.


Carcinoma, Hepatocellular , Liver Neoplasms , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Rodent Diseases , Alanine Transaminase/metabolism , Alanine Transaminase/pharmacology , Alanine Transaminase/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Aspartate Aminotransferases/metabolism , Aspartate Aminotransferases/pharmacology , Aspartate Aminotransferases/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/veterinary , Disease Models, Animal , Heterografts , Humans , Interleukin-10/metabolism , Interleukin-10/pharmacology , Interleukin-10/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/veterinary , Mesenchymal Stem Cell Transplantation/veterinary , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Sorafenib/metabolism , Sorafenib/pharmacology , Sorafenib/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
19.
J Feline Med Surg ; 24(8): e244-e250, 2022 08.
Article En | MEDLINE | ID: mdl-35713592

OBJECTIVES: The objective of this study was to compare the efficacy of feline mesenchymal stem cells (fMSC) with prednisolone as a treatment for inflammatory bowel disease (IBD) in cats. METHODS: Cats with chronic enteropathy that failed a 2-week diet trial and were not found to have significant concurrent disease were eligible for the study. If endoscopic biopsies confirmed a histopathologic diagnosis of IBD, the cat was randomly assigned to either the fMSC or prednisolone groups. Owners were blinded to the grouping. Stem cell treatment consisted of two intravenous injections of 2 × 106 cells/kg of freshly cultured allogeneic stem cells separated by 2 weeks. Prednisolone treatment was 1-2 mg/kg PO q24h, tapered according to clinical response. Owners were asked to make no changes (eg, diet and other medications) for the first 2 months, at which time they either continued to the 6-month recheck with no changes, or 'failed' treatment and owners were unblinded and changes made as necessary. RESULTS: Six prednisolone and six fMSC treatment cats completed the study. All six prednisolone group cats were spayed females with a mean age of 8.3 years (range 2-14), a mean body weight of 3.6 kg (range 2.5-4.8) and a mean pretreatment Feline Chronic Enteropathy Activity Index (FCEAI) score of 3.6 (range 2-6). The six stem cell cats included three spayed females and three castrated males, and had a mean age of 8.0 years (range 4.5-13), a mean body weight of 4.9 kg (range 4.0-5.9) and a mean pretreatment FCEAI score of 3.7 (range 2-5). One cat in each group failed at the 2-month recheck. At the 6-month recheck, the mean FCEAI score for the prednisolone group was 3.7 (range 0.5-9) and 0.75 (range 0-1.5) for the fMSC group. CONCLUSIONS AND RELEVANCE: These results suggest that this specific fMSC protocol appears to be as effective in the treatment of feline IBD as a standard course of prednisolone therapy.


Cat Diseases , Inflammatory Bowel Diseases , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Body Weight , Cat Diseases/drug therapy , Cats , Chronic Disease , Female , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/veterinary , Male , Mesenchymal Stem Cell Transplantation/veterinary , Prednisolone/therapeutic use
20.
Res Vet Sci ; 144: 157-163, 2022 May.
Article En | MEDLINE | ID: mdl-34802776

Mesenchymal stem cells are promising candidates for stem cell therapy in many diseases, especially in immune-associated diseases. Inflammatory bowel disease is a chronic autoimmune disease that can lead to colorectal cancer if it is not controlled. Mesenchymal stem cells are always under a hypoxic environment in vivo, whether in bone marrow or adipose tissue, whereas researchers always culture MSCs (mesenchymal stem cells) under normoxic conditions (21%). In this study, we aimed to investigate whether hypoxia (1%) affects the therapeutic effect of MSCs. We hypothesize that hypoxia may benefit the treatment efficacy of MSCs. We used DSS to induce IBD (inflammatory bowel disease) in mice and then injected MSCs that had been preconditioned under normoxic conditions (21%) and hypoxic conditions (1%). We found that compared with normoxic-preconditioned MSCs (n-MSCs), hypoxic-preconditioned MSCs (h-MSCs) could alleviate colon inflammation to a large extent, as determined by inflammatory cytokines and CD3+ T cell activation. Mechanistic studies showed that hypoxia could promote iNOS expression in MSCs. Therefore, our data suggest that hypoxia may be more appropriate than normoxia for facilitating MSCs exertion of therapeutic functions.


Colitis , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Rodent Diseases , Adipose Tissue , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/therapy , Colitis/veterinary , Hypoxia/metabolism , Hypoxia/veterinary , Mesenchymal Stem Cell Transplantation/veterinary , Mice
...