Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 27.594
Filtrer
1.
Stem Cell Res Ther ; 15(1): 204, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38978125

RÉSUMÉ

Spinal cord injury (SCI) is a complex tissue injury that results in a wide range of physical deficits, including permanent or progressive disabilities of sensory, motor and autonomic functions. To date, limitations in current clinical treatment options can leave SCI patients with lifelong disabilities. There is an urgent need to develop new therapies for reconstructing the damaged spinal cord neuron-glia network and restoring connectivity with the supraspinal pathways. Neural stem cells (NSCs) possess the ability to self-renew and differentiate into neurons and neuroglia, including oligodendrocytes, which are cells responsible for the formation and maintenance of the myelin sheath and the regeneration of demyelinated axons. For these properties, NSCs are considered to be a promising cell source for rebuilding damaged neural circuits and promoting myelin regeneration. Over the past decade, transplantation of NSCs has been extensively tested in a variety of preclinical models of SCI. This review aims to highlight the pathophysiology of SCI and promote the understanding of the role of NSCs in SCI repair therapy and the current advances in pathological mechanism, pre-clinical studies, as well as clinical trials of SCI via NSC transplantation therapeutic strategy. Understanding and mastering these frontier updates will pave the way for establishing novel therapeutic strategies to improve the quality of recovery from SCI.


Sujet(s)
Gaine de myéline , Cellules souches neurales , Traumatismes de la moelle épinière , Traumatismes de la moelle épinière/thérapie , Traumatismes de la moelle épinière/anatomopathologie , Humains , Cellules souches neurales/transplantation , Cellules souches neurales/cytologie , Gaine de myéline/métabolisme , Animaux , Régénération nerveuse/physiologie , Transplantation de cellules souches/méthodes
2.
Virol J ; 21(1): 149, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956615

RÉSUMÉ

OBJECTIVES: This study aimed to characterize incidences of CMV reactivations within one year post-allo-SCT and identify risk factors for CMV second reactivation episode in population with high seropositivity where first CMV reactivation episode deemed to be high. METHODS: This retrospective cohort study analyzed data from 359 allo-SCT patients aged 14 and older admitted to a tertiary academic hospital. Data on demographic and clinical factors, CMV serostatus, conditioning regimens, graft-versus-host disease prophylaxis, engraftment time, and CMV reactivations were collected. RESULTS: First and second CMV reactivations occurred in 88.9% and 18.4% of post-allo-SCT patients respectively. Patients were stratified into two groups based on primary disease necessitating allo-SCT, patients with malignant (Group 1) and non-malignant (Group 2) hematological disease. Factors associated with the second reactivation included cord blood as a stem cell source, human leukocyte antigen mismatch, acute graft-versus-host disease, and hematological malignancies. Patients with non-malignant hematological disease displayed better outcomes, including a higher rate of spontaneous clearance of first CMV reactivation (70% versus 49.4%) and lower rates of second CMV reactivation (9.6% versus 31%) than those with malignant hematological disease. The one-year overall survival rate was 87.7% (95.5% in non-malignant hematological disease and 78.13% in malignant hematological disease). CONCLUSION: Our findings are concordant with previous local study in regard to high rate of first CMV reactivation post-allo-SCT. It appears that patients with nonmalignant hematological disease had better outcomes, such as lower second CMV reactivation and higher survival rates compared to patients with malignant hematological disease. Further investigation is needed to identify other factors affecting recurrent CMV reactivations in allo-SCT in patients with malignant hematological disease.


Sujet(s)
Infections à cytomégalovirus , Cytomegalovirus , Transplantation homologue , Activation virale , Humains , Mâle , Femelle , Infections à cytomégalovirus/virologie , Infections à cytomégalovirus/épidémiologie , Adulte d'âge moyen , Adulte , Études rétrospectives , Jeune adulte , Cytomegalovirus/immunologie , Adolescent , Facteurs de risque , Sujet âgé , Transplantation homologue/effets indésirables , Récidive , Maladie du greffon contre l'hôte/étiologie , Transplantation de cellules souches hématopoïétiques/effets indésirables , Transplantation de cellules souches/effets indésirables , Incidence
3.
Stem Cell Res Ther ; 15(1): 212, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39020391

RÉSUMÉ

BACKGROUND: Sciatic nerve repair becomes a focus of research in neurological aspect to restore the normal physical ability of the animal to stand and walk. Tissue engineered nerve grafts (TENGs) provide a promising alternative therapy for regeneration of large gap defects. The present study investigates the regenerative capacity of PRP, ADSCs, and PRP mixed ADSCs on a long sciatic nerve defect (40-mm) bridged by a polyglycolic polypropylene (PGA-PRL) mesh which acts as a neural scaffold. MATERIALS AND METHODS: The study was conducted on 12 adult male mongrel dogs that were randomly divided into 4 groups: Group I (scaffold group); where the sciatic defect was bridged by a (PGA-PRL) mesh only while the mesh was injected with ADSCs in Group II (ADSCs group), PRP in Group III (PRP group). Mixture of PRP and ADSCs was allocated in Group IV (PRP + ADSCs group). Monthly, all animals were monitored for improvement in their gait and a numerical lameness score was recorded for all groups. 6 months-post surgery, the structural and functional recovery of sciatic nerve was evaluated electrophysiologically, and on the level of gene expression, and both sciatic nerve and the gastrocnemius muscle were evaluated morphometrically, histopathologically. RESULTS: Numerical lameness score showed improvement in the motor activities of both Group II and Group III followed by Group IV and the scaffold group showed mild improvement even after 6 months. Histopathologically, all treated groups showed axonal sprouting and numerous regenerated fascicles with obvious angiogenesis in proximal cut, and distal portion where Group IV exhibited a significant remyelination with the MCOOL technique. The regenerative ratio of gastrocnemius muscle was 23.81%, 56.68%, 52.06% and 40.69% for Group I, II, III and IV; respectively. The expression of NGF showed significant up regulation in the proximal portion for both Group III and Group IV (P ≤ 0.0001) while Group II showed no significant difference. PDGF-A, and VEGF expressions were up-regulated in Group II, III, and IV whereas Group I showed significant down-regulation for NGF, PDGF-A, and VEGF (P ≤ 0.0001). CONCLUSION: ADSCs have a great role in restoring the damaged nerve fibers by secreting several types of growth factors like NGF that have a proliferative effect on Schwann cells and their migration. In addition, PRP therapy potentiates the effect of ADSCs by synthesis another growth factors such as PDGF-A, VEGF, NGF for better healing of large sciatic gap defects.


Sujet(s)
Régénération nerveuse , Polypropylènes , Nerf ischiatique , Animaux , Chiens , Régénération nerveuse/physiologie , Nerf ischiatique/traumatismes , Mâle , Polypropylènes/composition chimique , Plasma riche en plaquettes/métabolisme , Tissu adipeux/cytologie , Acide polyglycolique/composition chimique , Cellules souches/cytologie , Cellules souches/métabolisme , Modèles animaux de maladie humaine , Structures d'échafaudage tissulaires/composition chimique , Transplantation de cellules souches/méthodes , Ingénierie tissulaire/méthodes
4.
Stem Cell Res Ther ; 15(1): 217, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39020406

RÉSUMÉ

BACKGROUND: Intrauterine adhesions (IUAs) jeopardise uterine function in women, which is a great challenge in the clinic. Previous studies have shown that endometrial perivascular cells (En-PSCs) can improve the healing of scarred uteri and that hydroxysafflor yellow A (HSYA) promotes angiogenesis. The purpose of this study was to observe whether the combination of En-PSCs with HSYA could improve the blood supply and fertility in the rat uterus after full-thickness injury. METHODS: En-PSCs were sorted by flow cytometry, and the effect of HSYA on the proliferation and angiogenesis of the En-PSCs was detected using CCK-8 and tube formation assays. Based on a previously reported rat IUA model, the rat uteri were sham-operated, spontaneously regenerated, or treated with collagen-loaded PBS, collagen-loaded HSYA, collagen-loaded En-PSCs, or collagen-loaded En-PSCs with HSYA, and then collected at both 30 and 90 days postsurgery. HE staining and Masson staining were used to evaluate uterine structure and collagen fibre deposition, and immunohistochemical staining for α-SMA and vWF was used to evaluate myometrial regeneration and neovascularization in each group. A fertility assay was performed to detect the recovery of pregnancy function in each group. RNA-seq was performed to determine the potential mechanism underlying En-PSCs/HSYA treatment. Immunofluorescence, tube formation assays, and Western blot were used to validate the molecular mechanism involved. RESULTS: The transplantation of Collagen/En-PSCs/HSYA markedly promoted uterine repair in rats with full-thickness injury by reducing fibrosis, increasing endometrial thickness, regenerating myometrium, promoting angiogenesis, and facilitated live births. RNA sequencing results suggested that En-PSCs/HSYA activated the NRG1/ErbB4 signaling pathway. In vitro tube formation experiments revealed that the addition of an ErbB inhibitor diminished the tube formation ability of cocultured En-PSCs and HUVECs. Western blot results further showed that elevated levels of NRG1 and ErbB4 proteins were detected in the Collagen/En-PSCs/HSYA group compared to the Collagen/En-PSCs group. These collective results suggested that the beneficial effects of the transplantation of Collagen/En-PSCs/HSYA might be attributed to the modulation of the NRG1/ErbB4 signaling pathway. CONCLUSIONS: The combination of En-PSCs/HSYA facilitated morphological and functional repair in rats with full-thickness uterine injury and may promote endometrial angiogenesis by regulating the NRG1/ErbB4 signaling pathway.


Sujet(s)
Chalcone , Endomètre , Quinones , Utérus , Animaux , Femelle , Rats , Endomètre/effets des médicaments et des substances chimiques , Endomètre/métabolisme , Humains , Utérus/effets des médicaments et des substances chimiques , Utérus/métabolisme , Chalcone/analogues et dérivés , Chalcone/pharmacologie , Quinones/pharmacologie , Quinones/usage thérapeutique , Rat Sprague-Dawley , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Cellules souches/métabolisme , Cellules souches/effets des médicaments et des substances chimiques , Transplantation de cellules souches/méthodes , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régénération/effets des médicaments et des substances chimiques
5.
PLoS One ; 19(7): e0304073, 2024.
Article de Anglais | MEDLINE | ID: mdl-38968328

RÉSUMÉ

BACKGROUND: Pharmacologic immunosuppression regimes are commonly employed in stem cell clinical trials to mitigate host immune rejection and promote survival and viability of transplanted cells. Immunosuppression and cell survival has been extensively studied in retinal and spinal tissues. The applicability of stem cell therapy is rapidly expanding to other sensory organs such as the ear and hearing. As regenerative therapy is directed to new areas, a greater understanding of immunosuppression strategies and their efficacy is required to facilitate translation to organ-specific biologic microenvironments. OBJECTIVE: This systematic review appraises the current literature regarding immunosuppression strategies employed in stem cell trials of retinal and neural cells. METHODS: This systematic review was performed in line with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Inclusion criteria included studies presenting data on neural or retinal cells as part of an in-human clinical trial that detailed the immunosuppression regime used. Exclusion criteria included non-English language studies, animal studies, review articles, case reports, editorials, and letters. The databases Medline, Embase, Scopus, Web of Science, and the Cochrane Library were searched from inception to February 2024. Risk of bias was evaluated using the ROBINS-I tool. RESULTS: Eighteen articles fit the inclusion criteria. Nine articles concerned retinal cells, 5 concerned spinal cord injury, and 4 concerned amyotrophic lateral sclerosis. A multi-drug and short-term immunosuppression regime were commonly employed in the identified studies. Detected immune responses in treated patients were rare. Common immunosuppression paradigms included tacrolimus, mycophenolate mofetil and tapering doses of steroids. Local immunosuppression with steroids was employed in some studies concerning retinal diseases. DISCUSSION: A short-term course of systemic immunosuppression seemed efficacious for most included studies, with some showing grafted cells viable months to years after immunosuppression had stopped. Longer-term follow-up is required to see if this remains the case. Side effects related to immunosuppression were uncommon.


Sujet(s)
Immunosuppression thérapeutique , Transplantation de cellules souches , Humains , Transplantation de cellules souches/méthodes , Immunosuppression thérapeutique/méthodes , Rétine/immunologie , Immunosuppresseurs/usage thérapeutique , Essais cliniques comme sujet
6.
Medicine (Baltimore) ; 103(27): e38768, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38968496

RÉSUMÉ

Antiretroviral therapy, also known as antiretroviral therapy (ART), has been at the forefront of the ongoing battle against human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDs). ART is effective, but it has drawbacks such as side effects, medication resistance, and difficulty getting access to treatment, which highlights the urgent need for novel treatment approaches. This review explores the complex field of HIV/AIDS treatment, covering both established alternative treatment modalities and orthodox antiretroviral therapy. Numerous reliable databases were reviewed, including PubMed, Web of Science, Scopus, and Google Scholar. The results of a thorough literature search revealed numerous therapeutic options, including stem cell transplantation, immunotherapy, gene therapy, latency reversal agents, and pharmaceutical vaccinations. While gene therapy has promise for altering cellular resistance to infection and targeting HIV-positive cells, immunotherapy treatments seek to strengthen the immune system's ability to combat HIV. Latency reversal agents offer a promising method of breaking the viral latency and making infected cells vulnerable to immune system destruction or antiretroviral drugs. Furthermore, there is potential for improving immune responses against HIV using medical vaccinations. This review stresses the vital significance of ongoing research and innovation in the hunt for a successful HIV/AIDS treatment through a thorough examination of recent developments and lingering challenges. The assessment notes that even though there has been tremendous progress in treating the illness, there is still more work to be done in addressing current barriers and investigating various treatment options in order to achieve the ultimate objective of putting an end to the HIV/AIDS pandemic.


Sujet(s)
Infections à VIH , Humains , Infections à VIH/traitement médicamenteux , Immunothérapie/méthodes , Syndrome d'immunodéficience acquise/traitement médicamenteux , Thérapie génétique , Latence virale/effets des médicaments et des substances chimiques , Antirétroviraux/usage thérapeutique , Agents antiVIH/usage thérapeutique , Transplantation de cellules souches
7.
Hum Vaccin Immunother ; 20(1): 2370970, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38977418

RÉSUMÉ

The impact of pre-graft COVID-19 vaccinations in donor or recipient as well as pre-graft infection has been studied in 157 adults having received allogeneic stem cell transplantation (Allo-SCT) for various hematological diseases during the delta/omicron waves. We showed here that pre-Allo-SCT COVID-19 vaccination and/or infection do not provide more protection in patients receiving vaccine, immunotherapy or both after transplant. COVID-19 vaccination is and remains of crucial importance after Allo-SCT, reinforcing the recommendation to start COVID-19 vaccination as soon as the third month following the transplant.


Sujet(s)
Vaccins contre la COVID-19 , COVID-19 , Transplantation de cellules souches hématopoïétiques , Transplantation homologue , Vaccination , Humains , COVID-19/prévention et contrôle , COVID-19/immunologie , Mâle , Vaccins contre la COVID-19/administration et posologie , Vaccins contre la COVID-19/immunologie , Femelle , Adulte d'âge moyen , Adulte , Transplantation homologue/effets indésirables , Transplantation de cellules souches hématopoïétiques/effets indésirables , Sujet âgé , Immunothérapie/méthodes , SARS-CoV-2/immunologie , Transplantation de cellules souches/effets indésirables
8.
Microsurgery ; 44(5): e31208, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39012167

RÉSUMÉ

BACKGROUND: Traumatic peripheral nerve injury, with an annual incidence reported to be approximately 13-23 per 100,000 people, is a serious clinical condition that can often lead to significant functional impairment and permanent disability. Although nerve transfer has become increasingly popular in the treatment of brachial plexus injuries, satisfactory results cannot be obtained even with total nerve root transfer, especially after serious injuries. To overcome this problem, we hypothesize that the application of stem cells in conjunction with nerve transfer procedures may be a viable alternative to more aggressive treatments that do not result in adequate improvement. Similarly, some preliminary studies have shown that adipose stem cells combined with acellular nerve allograft provide promising results in the repair of brachial plexus injury. The purpose of this study was to assess the efficacy of combining adipose-derived stem cells with nerve transfer procedure in a rat brachial plexus injury model. METHODS: Twenty female Wistar rats weighing 300-350 g and aged 8-10 weeks were randomly divided into two groups: a nerve transfer group (NT group) and a nerve transfer combined adipose stem cell group (NT and ASC group). The upper brachial plexus injury model was established by gently avulsing the C5-C6 roots from the spinal cord with microforceps. A nerve transfer from the ulnar nerve to the musculocutaneous nerve (Oberlin procedure) was performed with or without seeded allogeneic adipose tissue-derived stem cells. Adipose tissue-derived stem cells at a rate of 2 × 106 cells were injected locally to the surface of the nerve transfer area with a 23-gauge needle. Immunohistochemistry (S100 and PGP 9.5 antibodies) and electrophysiological data were used to evaluate the effect of nerve repair 12 weeks after surgery. RESULTS: The mean latency was significantly longer in the NT group (2.0 ± 0.0 ms, 95% CI: 1.96-2.06) than in the NT and ASC group (1.7 ± 0.0 ms, 95% CI: 1.7-1.7) (p < .001). The mean peak value was higher in the NT group (1.7 ± 0.0 mV, 95% CI: 1.7-1.7) than in the NT and ASC group (1.7 ± 0.3 mV, 95% CI: 1.6-1.9) with no significant difference (p = .61). Although S100 and PGP 9.5 positive areas were observed in higher amounts in the NT and ASC group compared to the NT group, the differences were not statistically significant (p = .26 and .08, respectively). CONCLUSIONS: This study conducted on rats provides preliminary evidence that adipose-derived stem cells may have a positive effect on nerve transfer for the treatment of brachial plexus injury. Further studies with larger sample sizes and longer follow-up periods are needed to confirm these findings.


Sujet(s)
Tissu adipeux , Plexus brachial , Modèles animaux de maladie humaine , Nerf musculocutané , Régénération nerveuse , Transfert nerveux , Rat Wistar , Nerf ulnaire , Animaux , Rats , Transfert nerveux/méthodes , Femelle , Régénération nerveuse/physiologie , Plexus brachial/traumatismes , Plexus brachial/chirurgie , Nerf musculocutané/chirurgie , Tissu adipeux/cytologie , Tissu adipeux/transplantation , Nerf ulnaire/traumatismes , Nerf ulnaire/chirurgie , Nerf ulnaire/transplantation , Transplantation de cellules souches/méthodes , Répartition aléatoire , Neuropathies du plexus brachial/chirurgie , Lésions des nerfs périphériques/chirurgie
9.
Mol Genet Genomic Med ; 12(6): e2479, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38860502

RÉSUMÉ

BACKGROUND: As a screening method, inaccuracies in noninvasive prenatal screening (NIPS) exist, which are often attributable to biological factors. One such factor is the history of transplantation. However, there are still limited reports on such NIPS cases. METHODS: We report an NIPS case of a pregnant woman who had received a stem cell transplant from a male donor. To determine the karyotype in the woman's original cell, we performed chromosome microarray analysis (CMA) on her postnatal blood and oral mucosa. To comprehensively estimate the cell-free DNA (cfDNA) composition, we further performed standard NIPS procedures on the postnatal plasma. Moreover, we reviewed all published relevant NIPS case reports about pregnant women with transplantation history. RESULTS: NIPS showed a low-risk result for common trisomies with a fetal fraction of 65.80%. CMA on maternal white blood cells showed a nonmosaic male karyotype, while the oral mucosa showed a nonmosaic female karyotype. The proportion of donor's cfDNA in postnatal plasma was 94.73% based on the Y-chromosome reads ratio. The composition of cfDNA in maternal plasma was estimated as follows: prenatally, 13.60% maternal, 65.80% donor, and 20.60% fetal/placental, whereas postnatally, 5.27% maternal and 94.73% donor. CONCLUSIONS: This study expanded our understanding of the influence of stem cell transplantation on NIPS, allowing us to optimize NIPS management for these women.


Sujet(s)
Acides nucléiques acellulaires , Dépistage prénatal non invasif , Humains , Femelle , Grossesse , Mâle , Adulte , Acides nucléiques acellulaires/génétique , Acides nucléiques acellulaires/sang , Dépistage prénatal non invasif/méthodes , Transplantation de cellules souches , Donneurs de tissus , Trisomie/génétique
10.
Stem Cell Res Ther ; 15(1): 163, 2024 Jun 09.
Article de Anglais | MEDLINE | ID: mdl-38853252

RÉSUMÉ

BACKGROUND: A rising population faces challenges with healing-impaired cutaneous wounds, often leading to physical disabilities. Adipose-derived stem cells (ASCs), specifically in the cell sheet format, have emerged as a promising remedy for impaired wound healing. Human platelet lysate (HPL) provides an attractive alternative to fetal bovine serum (FBS) for culturing clinical-grade ASCs. However, the potential of HPL sheets in promoting wound healing has not been fully investigated. This study aimed to explore the anti-fibrotic and pro-angiogenic capabilities of HPL-cultured ASC sheets and delve into the molecular mechanism. METHODS: A rat burn model was utilized to evaluate the efficacy of HPL-cultured ASC sheets in promoting wound healing. ASC sheets were fabricated with HPL, and those with FBS were included for comparison. Various analyses were conducted to assess the impact of HPL sheets on wound healing. Histological examination of wound tissues provided insights into aspects such as wound closure, collagen deposition, and overall tissue regeneration. Immunofluorescence was employed to assess the presence and distribution of transplanted ASCs after treatment. Further in vitro studies were conducted to decipher the specific factors in HPL sheets contributing to angiogenesis. RESULTS: HPL-cultured ASC sheets significantly accelerated wound closure, fostering ample and organized collagen deposition in the neo-dermis. Significantly more retained ASCs were observed in wound tissues treated with HPL sheets compared to the FBS counterparts. Moreover, HPL sheets mitigated macrophage recruitment and decreased subsequent wound tissue fibrosis in vivo. Immunohistochemistry also indicated enhanced angiogenesis in the HPL sheet group. The in vitro analyses showed upregulation of C-C motif chemokine ligand 5 (CCL5) and angiogenin in HPL sheets, including both gene expression and protein secretion. Culturing endothelial cells in the conditioned media compared to media supplemented with CCL5 or angiogenin suggested a correlation between CCL5 and the pro-angiogenic effect of HPL sheets. Additionally, through neutralizing antibody experiments, we further validated the crucial role of CCL5 in HPL sheet-mediated angiogenesis in vitro. CONCLUSIONS: The present study underscores CCL5 as an essential factor in the pro-angiogenic effect of HPL-cultured ASC sheets during the wound healing process. These findings highlight the potential of HPL-cultured ASC sheets as a promising therapeutic option for healing-impaired cutaneous wounds in clinical settings. Furthermore, the mechanism exploration yields valuable information for optimizing regenerative strategies with ASC products. BRIEF ACKNOWLEDGMENT: This research was supported by the National Science and Technology Council, Taiwan (NSTC112-2321-B-002-018), National Taiwan University Hospital (111C-007), and E-Da Hospital-National Taiwan University Hospital Joint Research Program (111-EDN0001, 112-EDN0002).


Sujet(s)
Tissu adipeux , Plaquettes , Chimiokine CCL5 , Néovascularisation physiologique , Cicatrisation de plaie , Animaux , Humains , Rats , Plaquettes/métabolisme , Chimiokine CCL5/métabolisme , Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Cellules souches/métabolisme , Cellules souches/cytologie , Rat Sprague-Dawley , Cellules cultivées , Mâle , Transplantation de cellules souches/méthodes ,
11.
Low Urin Tract Symptoms ; 16(4): e12527, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38867432

RÉSUMÉ

Interstitial cystitis/bladder pain syndrome (IC/BPS) is a chronic disease with limited treatment options. Current multidisciplinary approach targeting bladder inflammation and urothelial dysfunction has limited durable effect that major surgery is ultimately required for both Hunner and non-Hunner type IC. Various investigational attempts are underway to avoid such operations and preserve the urinary bladder. Stem cell therapy is a fascinating option for treating chronic illnesses. Stem cells can self-renew, restore damaged tissue, and have paracrine effects. The therapeutic efficacy and safety of stem cell therapy have been demonstrated in numerous preclinical models, primarily chemically induced cystitis rat models. Only one clinical trial (phase 1 study) has investigated the safety of human embryonic stem cell-derived mesenchymal stem cells in three Hunner-type IC patients. Under general anesthesia, participants underwent cystoscopic submucosal stem cell injection (2.0 × 107 stem cells/5 mL). No safety issues were reported up to 12 months of follow-up and long-term follow-up (up to 3 years). Although there were variations in therapeutic response, all patients reported significant improvement in pain at 1 month postoperatively. One patient underwent fulguration of the Hunner lesion after the trial, but others reported an overall improvement in pain. The analysis on phase 1/2a trial which had several modifications in protocol is currently ongoing. Despite several limitations that need to be overcome, stem cell therapy could be a potential therapeutic option for treating IC/BPS. Clinical outcome on phase 1/2a trial is important and might provide more insight into the clinical application of stem cell therapy for IC/BPS.


Sujet(s)
Cystite interstitielle , Transplantation de cellules souches , Cystite interstitielle/thérapie , Humains , Transplantation de cellules souches/méthodes , Animaux , Transplantation de cellules souches mésenchymateuses/méthodes
12.
Asian Pac J Cancer Prev ; 25(6): 2099-2104, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38918672

RÉSUMÉ

BACKGROUND AND OBJECTIVES: Recent studies have highlighted the potential of fetal hepatic stem cells in regenerative treatments for liver diseases. This study aimed to evaluate the short-term effects of fetal stem cell transplantation in patients with liver cirrhosis resulting from chronic hepatitis C. MATERIALS AND METHODS: Thirty patients with liver cirrhosis of all Child-Turcotte-Pugh classes due to chronic hepatitis C, aged 18 to 65 years, were selected for this study. A single intravenous dose of 1 ml containing 6*106 fetal hepatic stem cells, diluted in 20.0 ml of 0.9% sodium chloride solution, was administered. The efficacy of the treatment was assessed by measuring levels of ALT, AST, total and direct bilirubin, gamma-glutamyltranspeptidase, alkaline phosphatase, total protein, and albumin before and after cell therapy. RESULTS: Post-treatment, a significant reduction was noted in the Child-Pugh score from 8 [6-9] to 7 [6-8] (p<0.001) and the MELD index from 11 [7-15] to 10 [7-14] (p=0.004). Skin itching decreased from 36.7% to 10%. Complaints of weakness increased significantly from 3.3% to 23.3% after 30 days of therapy (p=0.014), and the incidence of reduced appetite increased from 20% to 46.7% (p=0.021). No statistical differences were observed in the frequency of nosebleeds (86.7% initially vs. 90% at day 30, p=0.655) or drowsiness (63.3% initially vs. 76.7% at day 30, p=0.157). Significant reductions were noted in ALT levels by 35% and total bilirubin by 44%. The lack of significant changes in indicators of hepatic-cell insufficiency, particularly the protein-forming function as reflected in total protein and albumin levels, is likely due to the extent of liver tissue damage and thus a delayed recovery. CONCLUSION: The findings of this study affirm the clinical efficacy and promise of fetal hepatic stem cell therapy as part of a comprehensive treatment regimen for patients with liver cirrhosis.


Sujet(s)
Hépatite C chronique , Cirrhose du foie , Humains , Cirrhose du foie/thérapie , Adulte d'âge moyen , Mâle , Femelle , Adulte , Adolescent , Hépatite C chronique/complications , Jeune adulte , Sujet âgé , Hepacivirus , Transplantation de cellules souches/méthodes , Études de suivi , Pronostic
13.
J Vis Exp ; (208)2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38912769

RÉSUMÉ

Traumatic spinal cord injury (SCI) induces permanent sensorimotor deficit below the site of injury. It affects approximately a quarter million people in the US, and it represents an immeasurable public health concern. Research has been conducted to provide effective therapy; however, SCI is still considered incurable due to the complex nature of the injury site. A variety of strategies, including drug delivery, cell transplantation, and injectable biomaterials, are investigated, but one strategy alone limits their efficacy for regeneration. As such, combinatorial therapies have recently gained attention that can target multifaceted features of the injury. It has been shown that extracellular matrices (ECM) may increase the efficacy of cell transplantation for SCI. To this end, 3D hydrogels consisting of decellularized spinal cords (dSCs) and sciatic nerves (dSNs) were developed at different ratios and characterized. Histological analysis of dSCs and dSNs confirmed the removal of cellular and nuclear components, and native tissue architectures were retained after decellularization. Afterward, composite hydrogels were created at different volumetric ratios and subjected to analyses of turbidity gelation kinetics, mechanical properties, and embedded human adipose-derived stem cell (hASC) viability. No significant differences in mechanical properties were found among the different ratios of hydrogels and decellularized spinal cord matrices. Human ASCs embedded in the gels remained viable throughout the 14-day culture. This study provides a means of generating tissue-engineered combinatorial hydrogels that present nerve-specific ECM and pro-regenerative mesenchymal stem cells. This platform can provide new insights into neuro-regenerative strategies after SCI with future investigations.


Sujet(s)
Hydrogels , Traumatismes de la moelle épinière , Traumatismes de la moelle épinière/thérapie , Hydrogels/composition chimique , Humains , Animaux , Transplantation de cellules souches/méthodes , Nerf ischiatique/traumatismes , Moelle spinale , Cellules souches mésenchymateuses/cytologie
14.
Tissue Cell ; 88: 102429, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38833939

RÉSUMÉ

Neuronal injuries, as one of the consequences of sports-related incidents, exert a profound influence on the athletes' future, potentially leading to complete immobility and impeding their athletic pursuits. In cases of severe damage inflicted upon the spinal cord (SC) and peripheral nervous systems (PNS), the regenerative process is notably compromised, rendering it essentially inefficient. Among the pivotal therapeutic approaches for the enhancement and prevention of secondary SC injuries (SCI), stem cell transplantation (SCT) stands out prominently. Stem cells, whether directly involved in replacement and reconstruction or indirectly through modification and secretion of crucial bioenvironmental factors, engage in the intricate process of tissue regeneration. Stem cells, through the secretion of neurotrophic factors (NTFs) (aiming to modulate the immune system), reduction of inflammation, axonal growth stimulation, and myelin formation, endeavor to facilitate the regeneration of damaged SC tissue. The fundamental challenges of this approach encompass the proper selection of suitable stem cell candidates for transplantation and the establishment of an appropriate microenvironment conducive to SC repair. In this article, an attempt has been made to explore sports-related injuries, particularly SCI, to comprehensively review innovative methods for treating SCI, and to address the existing challenges. Additionally, some of the stem cells used in neural injuries and the process of their utilization have been discussed.


Sujet(s)
Traumatismes sportifs , Traumatismes de la moelle épinière , Transplantation de cellules souches , Humains , Traumatismes de la moelle épinière/thérapie , Transplantation de cellules souches/méthodes , Traumatismes sportifs/thérapie , Animaux , Régénération nerveuse/physiologie , Système nerveux périphérique/traumatismes
15.
Cells ; 13(12)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38920674

RÉSUMÉ

Bone/fracture healing is a complex process with different steps and four basic tissue layers being affected: cortical bone, periosteum, fascial tissue surrounding the fracture, and bone marrow. Stem cells and their derivatives, including embryonic stem cells, induced pluripotent stem cells, mesenchymal stem cells, hematopoietic stem cells, skeletal stem cells, and multipotent stem cells, can function to artificially introduce highly regenerative cells into decrepit biological tissues and augment the healing process at the tissue level. Stem cells are molecularly and functionally indistinguishable from standard human tissues. The widespread appeal of stem cell therapy lies in its potential benefits as a therapeutic technology that, if harnessed, can be applied in clinical settings. This review aims to establish the molecular pathophysiology of bone healing and the current stem cell interventions that disrupt or augment the bone healing process and, finally, considers the future direction/therapeutic options related to stem cells and bone healing.


Sujet(s)
Régénération osseuse , Consolidation de fracture , Humains , Régénération osseuse/physiologie , Animaux , Cellules souches/cytologie , Transplantation de cellules souches/méthodes
16.
Cells ; 13(12)2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38920693

RÉSUMÉ

Bone tissue injuries within oral and dental contexts often present considerable challenges because traditional treatments may not be able to fully restore lost or damaged bone tissue. Novel approaches involving stem cells and targeted 3D scaffolds have been investigated in the search for workable solutions. The use of scaffolds in stem cell-assisted bone regeneration is a crucial component of tissue engineering techniques designed to overcome the drawbacks of traditional bone grafts. This study provides a detailed review of scaffold applications for bone regeneration with stem cells in dentistry. This review focuses on scaffolds and stem cells while covering a broad range of studies explaining bone regeneration in dentistry through the presentation of studies conducted in this field. The role of different stem cells in regenerative medicine is covered in great detail in the reviewed literature. These studies have addressed a wide range of subjects, including the effects of platelet concentrates during dental surgery or specific combinations, such as human dental pulp stem cells with scaffolds for animal model bone regeneration, to promote bone regeneration in animal models. Noting developments, research works consider methods to improve vascularization and explore the use of 3D-printed scaffolds, secretome applications, mesenchymal stem cells, and biomaterials for oral bone tissue regeneration. This thorough assessment outlines possible developments within these crucial regenerative dentistry cycles and provides insights and suggestions for additional study. Furthermore, alternative creative methods for regenerating bone tissue include biophysical stimuli, mechanical stimulation, magnetic field therapy, laser therapy, nutritional supplements and diet, gene therapy, and biomimetic materials. These innovative approaches offer promising avenues for future research and development in the field of bone tissue regeneration in dentistry.


Sujet(s)
Régénération osseuse , Odontologie , Cellules souches , Ingénierie tissulaire , Structures d'échafaudage tissulaires , Humains , Structures d'échafaudage tissulaires/composition chimique , Animaux , Cellules souches/cytologie , Odontologie/méthodes , Ingénierie tissulaire/méthodes , Pulpe dentaire/cytologie , Transplantation de cellules souches/méthodes , Médecine régénérative/méthodes
17.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38928214

RÉSUMÉ

Stroke is the number one cause of morbidity in the United States and number two cause of death worldwide. There is a critical unmet medical need for more effective treatments of ischemic stroke, and this need is increasing with the shift in demographics to an older population. Recently, several studies have reported the therapeutic potential of stem cell-derived exosomes as new candidates for cell-free treatment in stoke. This review focuses on the use of stem cell-derived exosomes as a potential treatment tool for stroke patients. Therapy using exosomes can have a clear clinical advantage over stem cell transplantation in terms of safety, cost, and convenience, as well as reducing bench-to-bed latency due to fewer regulatory milestones. In this review article, we focus on (1) the therapeutic potential of exosomes in stroke treatment, (2) the optimization process of upstream and downstream production, and (3) preclinical application in a stroke animal model. Finally, we discuss the limitations and challenges faced by exosome therapy in future clinical applications.


Sujet(s)
Exosomes , Accident vasculaire cérébral , Exosomes/métabolisme , Exosomes/transplantation , Humains , Animaux , Accident vasculaire cérébral/thérapie , Accident vasculaire cérébral/métabolisme , Transplantation de cellules souches/méthodes , Modèles animaux de maladie humaine , Cellules souches/métabolisme , Cellules souches/cytologie
18.
Life Sci ; 351: 122816, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38862064

RÉSUMÉ

AIMS: Parkinson's disease (PD) remains a substantial clinical challenge due to the progressive loss of midbrain dopaminergic (DA) neurons in nigrostriatal pathway. In this study, human amniotic epithelial stem cells (hAESCs)-derived dopaminergic neuron-like cells (hAESCs-DNLCs) were generated, with the aim of providing new therapeutic approach to PD. MATERIALS AND METHODS: hAESCs, which were isolated from discarded placentas, were induced to differentiate into hAESCs-DNLCs by following a "two stages" induction protocol. The differentiation efficiency was assessed by quantitative real-time PCR (qRT-PCR), immunocytochemistry (ICC), and ELISA. Immunogenicity, cell viability and tumorigenicity of hAESCs-DNLC were analyzed before in vivo experiments. Subsequently, hAESCs-DNLCs were transplanted into PD rats, behavioral tests were monitored after graft, and the regeneration of DA neurons was detected by immunohistochemistry (IHC). Furthermore, to trace hAESCs-DNLCs in vivo, cells were pre-labeled with PKH67 green fluorescence. KEY FINDINGS: hAESCs were positive for pluripotent markers and highly expressed neural stem cells (NSCs) markers. Based on this, we established an induction method reliably generates hAESCs-DNLCs, which was evidenced by epithelium-to-neuron morphological changes, elevated expressions of neuronal and DA neuronal markers, and increased secretion of dopamine. Moreover, hAESCs-DNLCs maintained high cell viability, no tumorigenicity and low immunogenicity, suggesting hAESCs-DNLCs an attractive implant for PD therapy. Transplantation of hAESCs-DNLCs into PD rats significantly ameliorated motor disorders, as well as enhanced the reinnervation of TH+ DA neurons in nigrostriatal pathway. SIGNIFICANCE: Our study has demonstrated evident therapeutic effects of hAESCs-DNLCs, and provides a safe and promising solution for PD.


Sujet(s)
Amnios , Différenciation cellulaire , Neurones dopaminergiques , Maladie de Parkinson , Rat Sprague-Dawley , Animaux , Neurones dopaminergiques/métabolisme , Rats , Humains , Amnios/cytologie , Maladie de Parkinson/thérapie , Femelle , Cellules épithéliales/métabolisme , Modèles animaux de maladie humaine , Mâle , Cellules souches neurales/transplantation , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Grossesse , Transplantation de cellules souches/méthodes , Cellules cultivées
19.
Life Sci ; 351: 122858, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38909681

RÉSUMÉ

Finding novel treatments for cardiovascular diseases (CVDs) is a hot topic in medicine; cell-based therapies have reported promising news for controlling dangerous complications of heart disease such as myocardial infarction (MI) and heart failure (HF). Various progenitor/stem cells were tested in various in-vivo, in-vitro, and clinical studies for regeneration or repairing the injured tissue in the myocardial to accelerate the healing. Fetal, adult, embryonic, and induced pluripotent stem cells (iPSC) have revealed the proper potency for cardiac tissue repair. As an essential communicator among cells, exosomes with specific contacts (proteins, lncRNAs, and miRNAs) greatly promote cardiac rehabilitation. Interestingly, stem cell-derived exosomes have more efficiency than stem cell transplantation. Therefore, stem cells induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), cardiac stem cells (CDC), and skeletal myoblasts) and their-derived exosomes will probably be considered an alternative therapy for CVDs remedy. In addition, stem cell-derived exosomes have been used in the diagnosis/prognosis of heart diseases. In this review, we explained the advances of stem cells/exosome-based treatment, their beneficial effects, and underlying mechanisms, which will present new insights in the clinical field in the future.


Sujet(s)
Exosomes , Défaillance cardiaque , Régénération , Transplantation de cellules souches , Humains , Exosomes/métabolisme , Exosomes/transplantation , Exosomes/physiologie , Défaillance cardiaque/thérapie , Défaillance cardiaque/métabolisme , Transplantation de cellules souches/méthodes , Animaux , Régénération/physiologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Cellules souches/métabolisme
20.
Int J Mol Sci ; 25(11)2024 May 26.
Article de Anglais | MEDLINE | ID: mdl-38891960

RÉSUMÉ

Heart failure (HF) is a life-threatening disorder and is treated by drug therapies and surgical interventions such as heart transplantation and left ventricular assist device (LVAD). However, these treatments can lack effectiveness in the long term and are associated with issues such as donor shortage in heart transplantation, and infection, stroke, or gastrointestinal bleeding in LVADs. Therefore, alternative therapeutic strategies are still needed. In this respect, stem cell therapy has been introduced for the treatment of HF and numerous preclinical and clinical studies are employing a range of stem cell varieties. These stem cells, such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), have been shown to improve cardiac function and attenuate left ventricular remodeling. IPSCs, which have a capacity for unlimited proliferation and differentiation into cardiomyocytes, are a promising cell source for myocardial regeneration therapy. In this review, we discuss the following topics: (1) what are iPSCs; (2) the limitations and solutions for the translation of iPSC-CMs practically; and (3) the current therapeutic clinical trials.


Sujet(s)
Défaillance cardiaque , Cellules souches pluripotentes induites , Myocytes cardiaques , Médecine régénérative , Humains , Cellules souches pluripotentes induites/cytologie , Myocytes cardiaques/cytologie , Médecine régénérative/méthodes , Défaillance cardiaque/thérapie , Animaux , Régénération , Transplantation de cellules souches/méthodes , Différenciation cellulaire
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...