Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
1.
Eur J Med Res ; 27(1): 5, 2022 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-35022077

RESUMO

BACKGROUND: Intervertebral disc degeneration (IDD) is a natural progression of age-related processes. Associated with IDD, degenerative disc disease (DDD) is a pathologic condition implicated as a major cause of chronic lower back pain, which can have a severe impact on the quality of life of patients. As degeneration progression is associated with elevated levels of inflammatory cytokines, enhanced aggrecan and collagen degradation, and changes in the disc cell phenotype. The purpose of this study was to investigate the biological and cytological characteristics of rabbit nucleus pulposus mesenchymal stem cells (NPMSCs)-a key factor in IDD-and to determine the effect of the growth and differentiation factor-5 (GDF5) on the differentiation of rabbit NPMSCs transduced with a lentivirus vector. METHODS: An in vitro culture model of rabbit NPMSCs was established and NPMSCs were identified by flow cytometry (FCM) and quantitative real-time PCR (qRT-PCR). Subsequently, NPMSCs were randomly divided into three groups: a transfection group (the lentiviral vector carrying GDF5 gene used to transfect NPMSCs); a control virus group (the NPMSCs transfected with an ordinary lentiviral vector); and a normal group (the NPMSCs alone). FCM, qRT-PCR, and western blot (WB) were used to detect the changes in NPMSCs. RESULTS: The GDF5-transfected NPMSCs displayed an elongated shape, with decreased cell density, and significantly increased GDF5 positivity rate in the transfected group compared to the other two groups (P < 0.01). The mRNA levels of Krt8, Krt18, and Krt19 in the transfected group were significantly higher in comparison with the other two groups (P < 0.01), and the WB results were consistent with that of qRT-PCR. CONCLUSIONS: GDF5 could induce the differentiation of NPMSCs. The lentiviral vector carrying the GDF5 gene could be integrated into the chromosome genome of NPMSCs and promoted differentiation of NPMSCs into nucleus pulposus cells. Our findings advance the development of feasible and effective therapies for IDD.


Assuntos
Regulação da Expressão Gênica , Fator 5 de Diferenciação de Crescimento/genética , Infecções por Lentivirus/virologia , Lentivirus , Células-Tronco Mesenquimais/citologia , Núcleo Pulposo/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Fator 5 de Diferenciação de Crescimento/biossíntese , Infecções por Lentivirus/metabolismo , Infecções por Lentivirus/patologia , Células-Tronco Mesenquimais/virologia , Núcleo Pulposo/patologia , Núcleo Pulposo/virologia , Coelhos
2.
PLoS Pathog ; 17(12): e1009600, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34936683

RESUMO

Kaposi's sarcoma (KS) is an angioproliferative and invasive tumor caused by Kaposi's sarcoma-associated herpesvirus (KSHV). The cellular origin of KS tumor cells remains contentious. Recently, evidence has accrued indicating that KS may arise from KSHV-infected mesenchymal stem cells (MSCs) through mesenchymal-to-endothelial transition (MEndT), but the transformation process has been largely unknown. In this study, we investigated the KSHV-mediated MEndT process and found that KSHV infection rendered MSCs incomplete endothelial lineage differentiation and formed hybrid mesenchymal/endothelial (M/E) state cells characterized by simultaneous expression of mesenchymal markers Nestin/PDGFRA/α-SAM and endothelial markers CD31/PDPN/VEGFR2. The hybrid M/E cells have acquired tumorigenic phenotypes in vitro and the potential to form KS-like lesions after being transplanted in mice under renal capsules. These results suggest a homology of KSHV-infected MSCs with Kaposi's sarcoma where proliferating KS spindle-shaped cells and the cells that line KS-specific aberrant vessels were also found to exhibit the hybrid M/E state. Furthermore, the genetic analysis identified KSHV-encoded FLICE inhibitory protein (vFLIP) as a crucial regulator controlling KSHV-induced MEndT and generating hybrid M/E state cells for tumorigenesis. Overall, KSHV-mediated MEndT that transforms MSCs to tumorigenic hybrid M/E state cells driven by vFLIP is an essential event in Kaposi's sarcomagenesis.


Assuntos
Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Nestina/metabolismo , Sarcoma de Kaposi/virologia , Proteínas Virais/metabolismo , Animais , Carcinogênese , Diferenciação Celular , Células Endoteliais/patologia , Células Endoteliais/virologia , Feminino , Infecções por Herpesviridae/patologia , Herpesvirus Humano 8/fisiologia , Humanos , Células-Tronco Mesenquimais/patologia , Células-Tronco Mesenquimais/virologia , Camundongos , Nestina/genética , Sarcoma de Kaposi/patologia , Proteínas Virais/genética
3.
Int J Mol Sci ; 22(21)2021 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-34769246

RESUMO

The inflammatory response plays a central role in the complications of congenital pulmonary airway malformations (CPAM) and severe coronavirus disease 2019 (COVID-19). The aim of this study was to evaluate the transcriptional changes induced by SARS-CoV-2 exposure in pediatric MSCs derived from pediatric lung (MSCs-lung) and CPAM tissues (MSCs-CPAM) in order to elucidate potential pathways involved in SARS-CoV-2 infection in a condition of exacerbated inflammatory response. MSCs-lung and MSCs-CPAM do not express angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TRMPSS2). SARS-CoV-2 appears to be unable to replicate in MSCs-CPAM and MSCs-lung. MSCs-lung and MSCs-CPAM maintained the expression of stemness markers MSCs-lung show an inflammatory response (IL6, IL1B, CXCL8, and CXCL10), and the activation of Notch3 non-canonical pathway; this route appears silent in MSCs-CPAM, and cytokine genes expression is reduced. Decreased value of p21 in MSCs-lung suggested no cell cycle block, and cells did not undergo apoptosis. MSCs-lung appears to increase genes associated with immunomodulatory function but could contribute to inflammation, while MSCs-CPAM keeps stable or reduce the immunomodulatory receptors expression, but they also reduce their cytokines expression. These data indicated that, independently from their perilesional or cystic origin, the MSCs populations already present in a patient affected with CPAM are not permissive for SARS-CoV-2 entry, and they will not spread the disease in case of infection. Moreover, these MSCs will not undergo apoptosis when they come in contact with SARS-CoV-2; on the contrary, they maintain their staminality profile.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Anormalidades do Sistema Respiratório , SARS-CoV-2/fisiologia , Transcriptoma , COVID-19/genética , COVID-19/metabolismo , COVID-19/patologia , Estudos de Casos e Controles , Células Cultivadas , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Humanos , Lactente , Pulmão/anormalidades , Pulmão/metabolismo , Pulmão/patologia , Masculino , Células-Tronco Mesenquimais/patologia , Células-Tronco Mesenquimais/virologia , RNA-Seq , Anormalidades do Sistema Respiratório/genética , Anormalidades do Sistema Respiratório/patologia , Anormalidades do Sistema Respiratório/virologia
4.
Biol Open ; 10(9)2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34494647

RESUMO

Ex vivo, gene therapy is a powerful approach holding great promises for the treatment of both genetic and acquired diseases. Adeno-associated virus (AAV) vectors are a safe and efficient delivery system for modification of mesenchymal stem cells (MSC) that could maximize their therapeutic benefits. Assessment of MSC viability and functional activity after infection with new AAV serotypes is necessary, due to AAV tropism to specific cell types. We infected human and rat adipose-tissue MSC with hybrid AAV-DJ serotype vectors carrying GFP and SCF genes. GFP expression from AAV-DJ was about 1.5-fold superior to that observed with AAV-2 and lasted for at least 21 days as was evaluated by flow cytometry and fluorescence microscopy. AAV-DJ proves to be suitable for the infection of rat and human MSC with a similar efficiency. Infected MSC were still viable but showed a 25-30% growth-rate slowdown. Moreover, we found an increase of SERPINB2 mRNA expression in human MSC while expression of other oxidative stress markers and extracellular matrix proteins was not affected. These results suggest that there is a differential cellular response in MSC infected with AAV viral vectors, which should be taken into account as it can affect the expected outcome for the therapeutic application.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/sangue , Células-Tronco Mesenquimais/virologia , Proteínas Virais/sangue , Animais , Proteínas de Fluorescência Verde/metabolismo , Humanos , Ratos , Sorogrupo , Fator de Células-Tronco/metabolismo , Tropismo Viral/genética
5.
PLoS Pathog ; 17(9): e1009847, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34492084

RESUMO

Increasing evidence suggests that Kaposi's sarcoma (KS) arises from Kaposi's sarcoma-associated herpesvirus (KSHV)-infected mesenchymal stem cells (MSCs) through mesenchymal-to-endothelial transition (MEndT). KSHV infection promotes MSC differentiation of endothelial lineage and acquisition of tumorigeneic phenotypes. To understand how KSHV induces MEndT and transforms MSCs to KS cells, we investigated the mechanism underlying KSHV-mediated MSC endothelial lineage differentiation. Like embryonic stem cells, MSC differentiation and fate determination are under epigenetic control. Prospero homeobox 1 (PROX1) is a master regulator that controls lymphatic vessel development and endothelial differentiation. We found that the PROX1 gene in MSCs harbors a distinctive bivalent epigenetic signature consisting of both active marker H3K4me3 and repressive marker H3K27me3, which poises expression of the genes, allowing timely activation upon differentiation signals or environmental stimuli. KSHV infection effectively resolves the bivalent chromatin by decreasing H3K27me3 and increasing H3K4me3 to activate the PROX1 gene. vIL-6 signaling leads to the recruitment of MLL2 and SET1 complexes to the PROX1 promoter to increase H3K4me3, and the vGPCR-VEGF-A axis is responsible for removing PRC2 from the promoter to reduce H3K27me3. Therefore, through a dual signaling process, KSHV activates PROX1 gene expression and initiates MEndT, which renders MSC tumorigenic features including angiogenesis, invasion and migration.


Assuntos
Diferenciação Celular/fisiologia , Transformação Celular Viral/fisiologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Mesenquimais/virologia , Sarcoma de Kaposi/virologia , Proteínas Supressoras de Tumor/metabolismo , Regulação da Expressão Gênica , Herpesvirus Humano 8 , Humanos
6.
Viruses ; 13(6)2021 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-34204818

RESUMO

Human multipotent mesenchymal stromal cells (hMSCs) are currently developed as cell therapeutics for different applications, including regenerative medicine, immune modulation, and cancer treatment. The biological properties of hMSCs can be further modulated by genetic engineering. Viral vectors based on human adenovirus type 5 (HAdV-5) belong to the most frequently used vector types for genetic modification of human cells in vitro and in vivo. However, due to a lack of the primary attachment receptor coxsackievirus and adenovirus receptor (CAR) in hMSCs, HAdV-5 vectors are currently not suitable for transduction of this cell type without capsid modification. Here we present several transduction enhancers that strongly enhance HAdV-5-mediated gene transfer into both bone marrow- and adipose tissue-derived hMSCs. Polybrene, poly-l-lysine, human lactoferrin, human blood coagulation factor X, spermine, and spermidine enabled high eGFP expression levels in hMSCs. Importantly, hMSCs treated with enhancers were not affected in their migration behavior, which is a key requisite for many therapeutic applications. Exemplary, strongly increased expression of tumor necrosis factor (TNF)-stimulated gene 6 (TSG-6) (a secreted model therapeutic protein) was achieved by enhancer-facilitated HAdV-5 transduction. Thus, enhancer-mediated HAdV-5 vector transduction is a valuable method for the engineering of hMSCs, which can be further exploited for the development of innovative hMSC therapeutics.


Assuntos
Adenovírus Humanos/genética , Vetores Genéticos , Células-Tronco Mesenquimais/virologia , Transdução Genética/métodos , Diferenciação Celular , Células Cultivadas , Técnicas de Cocultura , Terapia Genética/métodos , Humanos , Macrófagos/fisiologia
7.
J Cell Biochem ; 122(10): 1360-1375, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34056765

RESUMO

Cancer has remained a major health problem around the world. Mesenchymal stem cells (MSCs)-based therapy exhibits a therapeutic effect via different mechanisms. By using MSCs as carrier cells, the major problem of clearance of oncolytic viruses is resolved by neutralizing antibodies before they react with cancer cells. The aim of this study was to characterize the effect of infected MSCs by reovirus type-3 Dearing (T3D) for in vitro cancer therapy. Adipose-derived MSCs (AD-MSCs) were infected with reovirus T3D and its biological properties were evaluated. Then, the effects of reovirus-infected AD-MSCs on cytokine profile, nitric oxide (NO) production, and apoptosis induction in TC-1 cells were assessed. Our results indicated that the differentiation potential of AD-MSCs was affected by reovirus. However, phenotypes were not affected after infection. Then, the effects of reovirus-infected AD-MSCs in TC-1 cells showed an increased amount of tumor necrosis factor-alpha (TNF-α) and NO production and a decreased amount of transforming growth factor-beta 1 (TGF-ß1) and interleukin-10 (IL-10). Moreover, apoptosis significantly increased via coculturing of TC-1 cells with infected AD-MSCs, compared with control, and both internal and external apoptosis pathways are activated in experimental groups. In conclusion, the data showed that with increasing TNF-α and NO production and reducing IL-10 and TGF-ß production, AD-MSCs can enhance the oncolytic effect of reovirus in cancer cells. Furthermore, the results suggested that AD-MSCs can be used as effective carrier cells candidate for reovirus T3D to maximize their anticancer cell activity.


Assuntos
Neoplasias Pulmonares/terapia , Células-Tronco Mesenquimais/citologia , Terapia Viral Oncolítica/métodos , Reoviridae/genética , Animais , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Feminino , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Células-Tronco Mesenquimais/virologia , Camundongos , Camundongos Endogâmicos C57BL
8.
Arch Virol ; 166(8): 2285-2289, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34057608

RESUMO

Mesenchymal stromal cells (MSCs) are considered multipotent progenitors with the capacity to differentiate into mesoderm-like cells in many species. The immunosuppressive properties of MSCs are important for downregulating inflammatory responses. Turkey coronavirus (TCoV) is the etiological agent of a poult mortality syndrome that affects intestinal epithelial cells. In this study, poult MSCs were isolated, characterized, and infected with TCoV after in vitro culture. The poult-derived MSCs showed fibroblast-like morphology and the ability to undergo differentiation into mesodermal-derived cells and to support virus replication. Infection with TCoV resulted in cytopathic effects and the loss of cell viability. TCoV antigens and new viral progeny were detected at high levels, as were transcripts of the pro-inflammatory factors INFγ, IL-6, and IL-8. These findings suggest that the cytokine storm phenomenon is not restricted to one genus of the family Coronaviridae and that MSCs cannot always balance the process.


Assuntos
Coronavirus do Peru/fisiologia , Citocinas/metabolismo , Replicação Viral , Animais , Diferenciação Celular , Sobrevivência Celular , Efeito Citopatogênico Viral , Interferon gama/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/virologia , Perus , Regulação para Cima
9.
Cancer Lett ; 509: 26-38, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33819529

RESUMO

Oncolytic adenovirus-mediated gene therapy shows promise for cancer treatment; however, the systemic delivery of oncolytic adenovirus to tumors remains challenging. Recently, mesenchymal stem cells (MSCs) have emerged as potential vehicles for improving delivery. Yet, because the oncolytic adenovirus replicates in MSCs, balancing MSC viability with viral load is key to achieving optimal therapeutic effect. We thus developed an all-in-one Tet-on system that can regulate replication of oncolytic adenovirus. Then, we loaded the novel oncolytic adenovirus carrying interleukin (IL)-24 and/or Endostatin in human umbilical cord blood-mesenchymal stem cells (hUCB-MSCs) for glioma therapy. In vitro assays demonstrated that this novel oncolytic adenovirus could efficiently replicate and kill glioma cells while sparing normal cells. Moreover, doxycycline effectively regulated oncolytic adenovirus replication in the hUCB-MSCs. The doxycycline induction group with dual expression of IL-24 and Endostatin exhibited significantly greater antitumor effects than other groups in a xenograft model of glioma. Thus, this strategy for systemic delivery of oncolytic adenovirus with its oncolytic activity controlled by a Tet-on system is a promising method for achieving antitumor efficacy in glioma, especially for metastatic tumors.


Assuntos
Neoplasias Encefálicas/terapia , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Endostatinas/biossíntese , Terapia Genética , Glioma/terapia , Interleucinas/biossíntese , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/virologia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/virologia , Morte Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Endostatinas/genética , Feminino , Vetores Genéticos , Glioma/genética , Glioma/metabolismo , Glioma/virologia , Humanos , Interleucinas/genética , Camundongos Endogâmicos BALB C , Camundongos Nus , Vírus Oncolíticos/crescimento & desenvolvimento , Carga Tumoral , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Am J Pathol ; 191(7): 1255-1268, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33887214

RESUMO

We postulate that similar to bacteria, adult stem cells may also exhibit an altruistic defense mechanism to protect their niche against external threat. Herein, we report mesenchymal stem cell (MSC)-based altruistic defense against a mouse model of coronavirus, murine hepatitis virus-1 (MHV-1) infection of lung. MHV-1 infection led to reprogramming of CD271+ MSCs in the lung to an enhanced stemness phenotype that exhibits altruistic behavior, as per previous work in human embryonic stem cells. The reprogrammed MSCs exhibited transient expansion for 2 weeks, followed by apoptosis and expression of stemness genes. The conditioned media of the reprogrammed MSCs exhibited direct antiviral activity in an in vitro model of MHV-1-induced toxicity to type II alveolar epithelial cells by increasing their survival/proliferation and decreasing viral load. Thus, the reprogrammed MSCs can be identified as altruistic stem cells (ASCs), which exert a unique altruistic defense against MHV-1. In a mouse model of MSC-mediated Mycobacterium tuberculosis (MTB) dormancy, MHV-1 infection in the lung exhibited 20-fold lower viral loads than the MTB-free control mice on the third week of viral infection, and exhibited six-fold increase of ASCs, thereby enhancing the altruistic defense. Notably, these ASCs exhibited intracellular replication of MTB, and their extracellular release. Animals showed tuberculosis reactivation, suggesting that dormant MTB may exploit ASCs for disease reactivation.


Assuntos
Pulmão/virologia , Células-Tronco Mesenquimais/virologia , SARS-CoV-2 , Tuberculose/virologia , Animais , Modelos Animais de Doenças , Camundongos , Vírus da Hepatite Murina
11.
J Immunother Cancer ; 9(3)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33737338

RESUMO

BACKGROUND: Osteosarcoma is the most common malignant solid tumor that affects bones, however, survival rates of patients with relapsed osteosarcoma have not improved in the last 30 years. Oncolytic virotherapy, which uses viruses designed to selectively replicate in cancer cells, has emerged as a promising treatment for solid tumors. Our group uses mesenchymal stem cells (MSCs) to transport oncolytic adenoviruses (OAds) to the tumor site, a therapeutic strategy called Celyvir. This treatment has been already applied in human patients, canine patients and different mouse models. In parallel, previous results have probed that administration of granulocyte-colony stimulating factor (G-CSF) increased immune infiltration in tumors. We then hypothesized that the mobilization of immune cells by G-CSF may increase the antitumor efficacy of Celyvir treatment by increasing the immune infiltration into the tumors. METHODS: In this study, we use a murine version of Celyvir consisting in murine MSCs carrying the murine OAd dlE102-here called OAd-MSCs-in an immunocompetent model of osteosarcoma. We tested the antitumoral efficacy of the combination of OAd-MSCs plus G-CSF. RESULTS: Our results show that treatment with OAd-MSCs or the union of OAd-MSCs with G-CSF (Combination) significantly reduced tumor growth of osteosarcoma in vivo. Moreover, treated tumors presented higher tumor infiltration of immune cells-especially tumor-infiltrating lymphocytes-and reduced T cell exhaustion, which seems to be enhanced in tumors treated with the Combination. The comparison of our results to those obtained from a cohort of pediatric osteosarcoma patients showed that the virotherapy induces immunological changes similar to those observed in patients with good prognosis. CONCLUSIONS: The results open the possibility of using cellular virotherapy for the treatment of bone cancers. Indeed, its combination with G-CSF may be considered for the improvement of the therapy.


Assuntos
Adenoviridae/patogenicidade , Neoplasias Ósseas/terapia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Agentes de Imunomodulação/farmacologia , Células-Tronco Mesenquimais/virologia , Terapia Viral Oncolítica , Vírus Oncolíticos/patogenicidade , Osteossarcoma/terapia , Adenoviridae/imunologia , Animais , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/patologia , Neoplasias Ósseas/virologia , Linhagem Celular Tumoral , Terapia Combinada , Efeito Citopatogênico Viral , Bases de Dados Genéticas , Humanos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Transplante de Células-Tronco Mesenquimais , Camundongos Endogâmicos BALB C , Vírus Oncolíticos/imunologia , Osteossarcoma/imunologia , Osteossarcoma/patologia , Osteossarcoma/virologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Microambiente Tumoral
12.
Mol Ther ; 29(5): 1808-1820, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33571680

RESUMO

The immunosuppressive tumor microenvironment (TME) is a formidable barrier to the success of adoptive cell therapies for solid tumors. Oncolytic immunotherapy with engineered adenoviruses (OAd) may disrupt the TME by infecting tumor cells, as well as surrounding stroma, to improve the functionality of tumor-directed chimeric antigen receptor (CAR)-T cells, yet efficient delivery of OAds to solid tumors has been challenging. Here we describe how mesenchymal stromal cells (MSCs) can be used to systemically deliver a binary vector containing an OAd together with a helper-dependent Ad (HDAd; combinatorial Ad vector [CAd]) that expresses interleukin-12 (IL-12) and checkpoint PD-L1 (programmed death-ligand 1) blocker. CAd-infected MSCs deliver and produce functional virus to infect and lyse lung tumor cells while stimulating CAR-T cell anti-tumor activity by release of IL-12 and PD-L1 blocker. The combination of this approach with administration of HER.2-specific CAR-T cells eliminates 3D tumor spheroids in vitro and suppresses tumor growth in two orthotopic lung cancer models in vivo. Treatment with CAd MSCs increases the overall numbers of human T cells in vivo compared to CAR-T cell only treatment and enhances their polyfunctional cytokine secretion. These studies combine the predictable targeting of CAR-T cells with the advantages of cancer cell lysis and TME disruption by systemic MSC delivery of oncolytic virotherapy: incorporation of immunostimulation by cytokine and checkpoint inhibitor production through the HDAd further enhances anti-tumor activity.


Assuntos
Anticorpos Monoclonais/genética , Dependovirus/fisiologia , Vírus Auxiliares/fisiologia , Interleucina-12/metabolismo , Neoplasias Pulmonares/terapia , Células-Tronco Mesenquimais/virologia , Receptores de Antígenos de Linfócitos T/metabolismo , Células A549 , Animais , Anticorpos Monoclonais/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Linhagem Celular Tumoral , Terapia Combinada , Dependovirus/genética , Vírus Auxiliares/genética , Humanos , Imunoterapia Adotiva , Interleucina-12/antagonistas & inibidores , Interleucina-12/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Terapia Viral Oncolítica , Receptor ErbB-2/imunologia , Microambiente Tumoral , Tropismo Viral , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Stem Cells ; 39(6): 707-722, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33586320

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic has grown to be a global public health crisis with no safe and effective treatments available yet. Recent findings suggest that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the coronavirus pathogen that causes COVID-19, could elicit a cytokine storm that drives edema, dysfunction of the airway exchange, and acute respiratory distress syndrome in the lung, followed by acute cardiac injury and thromboembolic events leading to multiorgan failure and death. Mesenchymal stem cells (MSCs), owing to their powerful immunomodulatory abilities, have the potential to attenuate the cytokine storm and have therefore been proposed as a potential therapeutic approach for which several clinical trials are underway. Given that intravenous infusion of MSCs results in a significant trapping in the lung, MSC therapy could directly mitigate inflammation, protect alveolar epithelial cells, and reverse lung dysfunction by normalizing the pulmonary microenvironment and preventing pulmonary fibrosis. In this review, we present an overview and perspectives of the SARS-CoV-2 induced inflammatory dysfunction and the potential of MSC immunomodulation for the prevention and treatment of COVID-19 related pulmonary disease.


Assuntos
COVID-19/imunologia , Síndrome da Liberação de Citocina/imunologia , Células-Tronco Mesenquimais/imunologia , SARS-CoV-2/imunologia , COVID-19/terapia , COVID-19/virologia , Síndrome da Liberação de Citocina/terapia , Síndrome da Liberação de Citocina/virologia , Humanos , Imunomodulação , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/virologia , Pandemias , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/terapia , Fibrose Pulmonar/virologia , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/terapia , Síndrome do Desconforto Respiratório/virologia , SARS-CoV-2/genética
14.
Stem Cell Rev Rep ; 17(2): 341-356, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33511519

RESUMO

In the end of 2019 COVID-19 emerged as a new threat worldwide and this disease present impaired immune system, exacerbated production of inflammatory cytokines, and coagulation disturbs. Mesenchymal stem cell (MSC) derived extracellular vesicles (EVs) have emerged as a therapeutic option due to its intrinsic properties to alleviate inflammatory responses, capable to promote the restoring of injured tissue. EVs contain heterogeneous cargo, including active microRNAs, small noncoding sequences involved in post-transcriptional gene repression or degradation and can attach in multiple targets. This study investigated whether the MSC-EVs miRNA cargo has the capacity to modulate the exacerbated cytokines, cell death and coagulation disturbs present in severe COVID-19. Through bioinformatics analysis, four datasets of miRNA, using different stem cell tissue sources (bone marrow, umbilical cord and adipose tissue), and one dataset of mRNA (bone marrow) were analyzed. 58 miRNAs overlap in the four miRNA datasets analyzed. Sequentially, those miRNAs present in at least two datasets, were analyzed using miRWalk for the 3'UTR binding target mRNA. The result predicted 258 miRNAs for exacerbated cytokines and chemokines, 266 miRNAs for cell death genes and 148 miRNAs for coagulation cascades. Some miRNAs may simultaneously attenuate inflammatory agents, inhibit cell death genes and key factors of coagulation cascade, consequently preventing tissue damage and coagulation disturbs. Therefore, the MSC-derived EVs due to their heterogeneous cargo are a potential multitarget approach able to improve the survival rates of severe COVID-19 patients.


Assuntos
COVID-19/imunologia , Vesículas Extracelulares/imunologia , Células-Tronco Mesenquimais/imunologia , MicroRNAs/imunologia , SARS-CoV-2/imunologia , Vesículas Extracelulares/virologia , Humanos , Células-Tronco Mesenquimais/virologia
15.
Stem Cells Transl Med ; 10(4): 568-571, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33237619

RESUMO

The use of mesenchymal stem cells (MSC) derived from several sources has been suggested as a major anti-inflammation strategy during the recent outbreak of coronavirus-19 (COVID-19). As the virus enters the target cells through the receptor ACE2, it is important to determine if the MSC population transfused to patients could also be a target for the virus entry. We report here that ACE2 is highly expressed in adult bone marrow, adipose tissue, or umbilical cord-derived MSC. On the other hand, placenta-derived MSC express low levels of ACE2 but only in early passages of cultures. MSC derived from human embryonic stem cell or human induced pluripotent stem cells express also very low levels of ACE2. The transcriptome analysis of the MSCs with lowest expression of ACE2 in fetal-like MSCs is found to be associated in particularly with an anti-inflammatory signature. These results are of major interest for designing future clinical MSC-based stem cell therapies for severe COVID-19 infections.


Assuntos
Enzima de Conversão de Angiotensina 2/imunologia , COVID-19/imunologia , Terapia Baseada em Transplante de Células e Tecidos , Células-Tronco Mesenquimais , SARS-CoV-2/imunologia , Transcriptoma/imunologia , Adulto , Feminino , Humanos , Recém-Nascido , Masculino , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/patologia , Células-Tronco Mesenquimais/virologia , Especificidade de Órgãos/imunologia
16.
Stem Cell Rev Rep ; 17(1): 4-8, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32399806

RESUMO

SARS-CoV-2 viruses are positive single-stranded RNA viruses, whose infection can be asymptomatic or lead to the coronavirus disease 2019 (Covid-19). Covid-19 is a respiratory infection with a significant impact on the hematopoietic system and hemostasis leading to several cardiovascular complications. Hematologic consequences of this new infection allowed medical community to start new treatment approaches concerning infection going from targeted anti-inflammatory drugs to anticoagulation or stem cell therapies. A better understanding of Covid-19 pathophysiology, in particular hematological disorders, will help to choose appropriate treatment strategies.


Assuntos
COVID-19/epidemiologia , Doenças Hematológicas/epidemiologia , SARS-CoV-2/patogenicidade , Trombose/epidemiologia , Coagulação Sanguínea/genética , COVID-19/sangue , COVID-19/patologia , COVID-19/virologia , Citocinas/genética , Doenças Hematológicas/sangue , Doenças Hematológicas/patologia , Doenças Hematológicas/virologia , Humanos , Inflamação/sangue , Inflamação/epidemiologia , Inflamação/patologia , Inflamação/virologia , Linfopenia/sangue , Linfopenia/epidemiologia , Linfopenia/virologia , Células-Tronco Mesenquimais/virologia , Trombose/sangue , Trombose/patologia , Trombose/virologia
17.
Stem Cell Reports ; 16(3): 419-427, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-32950067

RESUMO

Previous studies reported on the safety and applicability of mesenchymal stem/stromal cells (MSCs) to ameliorate pulmonary inflammation in acute respiratory distress syndrome (ARDS). Thus, multiple clinical trials assessing the potential of MSCs for COVID-19 treatment are underway. Yet, as SARS-inducing coronaviruses infect stem/progenitor cells, it is unclear whether MSCs could be infected by SARS-CoV-2 upon transplantation to COVID-19 patients. We found that MSCs from bone marrow, amniotic fluid, and adipose tissue carry angiotensin-converting enzyme 2 and transmembrane protease serine subtype 2 at low levels on the cell surface under steady-state and inflammatory conditions. We did not observe SARS-CoV-2 infection or replication in MSCs at steady state under inflammatory conditions, or in direct contact with SARS-CoV-2-infected Caco-2 cells. Further, indoleamine 2,3-dioxygenase 1 production in MSCs was not impaired in the presence of SARS-CoV-2. We show that MSCs are resistant to SARS-CoV-2 infection and retain their immunomodulation potential, supporting their potential applicability for COVID-19 treatment.


Assuntos
COVID-19/virologia , Inflamação/virologia , Células-Tronco Mesenquimais/virologia , SARS-CoV-2/patogenicidade , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/metabolismo , Células CACO-2 , Linhagem Celular Tumoral , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Inflamação/metabolismo , Células-Tronco Mesenquimais/metabolismo , Serina Endopeptidases/metabolismo , Tratamento Farmacológico da COVID-19
18.
Cancer Gene Ther ; 28(1-2): 64-73, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32457488

RESUMO

There is increasing evidence about the use of oncolytic adenoviruses (Ads) as promising immunotherapy agents. We have previously demonstrated the clinical efficiency of mesenchymal stem cells (MSCs) infected with oncolytic Ads as an antitumoral immunotherapy (called Celyvir) in human and canine patients, using ICOVIR-5 or ICOCAV17 as human and canine oncolytic Ads, respectively. Considering the better clinical outcomes of canine patients, in this study we searched for differences in cellular responses of human and canine MSCs to Ad infection that may help understand the mechanisms leading to higher antitumor immune response. We found that infection of human and canine MSCs with ICOVIR-5 or ICOCAV17 did not activate the NF-κB pathway or the interferon regulatory factors IRF3 and IRF7. However, we observed differences in the profile of cytokines secretion, as infection of canine MSCs with ICOCAV17 resulted in lower secretion of several cytokines. Moreover, we showed that infection of human MSCs with ICOVIR-5 increased the phosphorylation of a number of proteins, including AKT and c-JUN. Finally, we demonstrated that differences in regulation of AKT and c-JUN in human and canine MSCs by ICOVIR-5 or ICOCAV17 are intrinsic to each virus. Our findings suggest that ICOCAV17 induces a more limited host response in canine MSCs, which may be related to a better clinical outcome. This result opens the possibility to develop new human oncolytic Ads with these specific properties. In addition, this improvement could be imitated by selecting specific human MSC on the basis of a limited host response after Ad infection.


Assuntos
Adenoviridae/imunologia , Células-Tronco Mesenquimais/metabolismo , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Animais , Cães , Humanos , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/virologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-jun/imunologia
19.
Eur Rev Med Pharmacol Sci ; 24(20): 10839-10849, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33155245

RESUMO

OBJECTIVE: To investigate the influence of oncolytic reovirus on the biological activities of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) as a novel virotherapy strategy. MATERIALS AND METHODS: The Cell Counting Kit-8 assay was used to detect the viability of hUC-MSCs infected with different multiplicities of infection (MOIs) of reoviruses. The biological activities (proliferation, marker expression, multipotency, and migration) of hUC-MSCs were verified by assaying osteogenic and adipogenic differentiation potential, flow cytometry, and electrical cell-substrate impedance sensing, respectively. RESULTS: The viability of hUC-MSCs slightly decreased by infection with low titers of reoviruses. A MOI of 1 had no effect on the viability of hUC-MSCs within 96 h. The biological activities (proliferation, marker expression, multipotency, and migration) of hUC-MSCs were not affected by reovirus infection at a MOI of 1. CONCLUSIONS: Reovirus at a MOI of 1 had no effect on the biological activities of hUC-MSCs.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Reoviridae/metabolismo , Cordão Umbilical/metabolismo , Animais , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Células-Tronco Mesenquimais/virologia , Camundongos , Reoviridae/isolamento & purificação , Cordão Umbilical/virologia
20.
Stem Cell Res ; 49: 102066, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33242791

RESUMO

Due to the multi-potential differentiation and immunomodulatory function, mesenchymal stem cells (MSCs) have been widely used in the therapy of chronic and autoimmune diseases. Recently, the novel coronavirus disease 2019 (COVID-19) has grown to be a global public health emergency but no effective drug is available to date. Several studies investigated MSCs therapy for COVID-19 patients. However, it remains unclear whether MSCs could be the host cells of SARS-CoV-2 (severe acute respiratory syndrome coronavirus-2) and whether they might affect the SARS-CoV-2 entry into other cells. Here, we report that human MSCs barely express ACE2 and TMPRSS2, two receptors required for the virus endocytosis, indicating that MSCs are free from SARS-CoV-2 infection. Furthermore, we observed that MSCs were unable to induce the expression of ACE2 and TMPRSS2 in epithelial cells and macrophages. Importantly, under different inflammatory challenge conditions, implanted human MSCs failed to up-regulate the expression of ACE2 and TMPRSS2 in the lung tissues of mice. Intriguingly, we showed that a SARS-CoV-2 pseudovirus failed to infect MSCs and co-cultured MSCs did not increase the risk of SARS-CoV-2 pseudovirus infection in epithelial cells. All these results suggest that human MSCs have no risk of assisting SARS-CoV-2 infection and the use of MSCs as the therapy for COVID-19 patients is feasible and safe.


Assuntos
COVID-19/transmissão , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , SARS-CoV-2/metabolismo , Segurança , Enzima de Conversão de Angiotensina 2/biossíntese , Animais , Linhagem Celular , Xenoenxertos , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Células-Tronco Mesenquimais/virologia , Camundongos , Serina Endopeptidases/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...