Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Immunother Cancer ; 12(4)2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38684370

RESUMEN

BACKGROUND: Chimeric antigen receptor (CAR)-T cell quality and stemness are associated with responsiveness, durability, and memory formation, which benefit clinical responses. Autologous T cell starting material across patients with cancer is variable and CAR-T expansion or potency can fail during manufacture. Thus, strategies to develop allogeneic CAR-T platforms including the identification and expansion of T cell subpopulations that correspond with CAR-T potency are an active area of investigation. Here, we compared CAR-T cells generated from healthy adult peripheral blood T cells versus placental circulating T (P-T) cells. METHODS: CAR-T cells from healthy adult peripheral blood mononuclear cells (PBMCs) and P-T cells were generated using the same protocol. CAR-T cells were characterized in detail by a combination of multiparameter flow cytometry, functional assays, and RNA sequencing. In vivo antitumor efficacy and persistence of CAR-T cells were evaluated in a Daudi lymphoma xenograft model. RESULTS: P-T cells possess stemness advantages compared with T cells from adult PBMCs. P-T cells are uniformly naïve prior to culture initiation, maintain longer telomeres, resist immune checkpoint upregulation, and resist further differentiation compared with PBMC T cells during CD19 CAR-T manufacture. P-T CD19 CAR-T cells are equally cytotoxic as PBMC-CD19 CAR-T cells but produce less interferon gamma in response to lymphoma. Transcriptome analysis shows P-T CD19 CAR-T cells retain a stem-like gene signature, strongly associate with naïve T cells, an early memory phenotype, and a unique CD4 T cell signature compared with PBMC-CD19 CAR-T cells, which enrich for exhaustion and stimulated memory T cell signatures. Consistent with functional data, P-T CD19 CAR-T cells exhibit attenuated inflammatory cytokine and chemokine gene signatures. In a murine in vivo model, P-T CD19 CAR-T cells eliminate lymphoma beyond 90 days. PBMC-CD19 CAR-T cells provide a non-durable benefit, which only delays disease onset. CONCLUSION: We identified characteristics of T cell stemness enriched in P-T CD19 CAR-T which are deficient in PBMC-derived products and translate into response durability in vivo. Our findings demonstrate that placental circulating T cells are a valuable cell source for allogeneic CAR-T products. Stemness advantages inherent to P-T cells translate to in vivo persistence advantages and long-term durable activity.


Asunto(s)
Citocinas , Inmunoterapia Adoptiva , Leucocitos Mononucleares , Placenta , Receptores Quiméricos de Antígenos , Linfocitos T , Humanos , Femenino , Animales , Ratones , Embarazo , Placenta/inmunología , Placenta/metabolismo , Citocinas/metabolismo , Inmunoterapia Adoptiva/métodos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Adulto , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Arthritis Res Ther ; 25(1): 229, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-38017556

RESUMEN

OBJECTIVE: Human placenta-derived exosomes (pExo) were generated, characterized, and evaluated as a therapeutic candidate for the treatment of osteoarthritis (OA). METHODS: pExo was generated from full-term human placenta tissues by sequential centrifugation, purification, and sterile filtration. Upon analysis of particle size, cytokine composition, and exosome marker expression, pExo was further tested in cell-based assays to examine its effects on human chondrocytes. In vivo therapeutic efficacies were evaluated in a medial meniscal tear/medial collateral ligament tear (MCLT + MMT) rat model, in which animals received pExo injections intraarticularly and weight bearing tests during in-life stage while histopathology and immunohistochemistry were performed as terminal endpoints. RESULTS: pExo displayed typical particle size, expressed maker proteins of exosome, and contained proteins with pro-proliferative, pro-anabolic, anti-catabolic, or anti-inflammatory activities. In vitro, pExo promoted chondrocyte migration and proliferation dose-dependently, which may involve its activation of cell growth-related signaling pathways. Expression of inflammatory and catabolic genes induced in a cellular OA model was significantly suppressed by pExo. In the rat OA model, pExo alleviated pain burden, restored cartilage degeneration, and downregulated expressions of pro-inflammatory, catabolic, or apoptotic proteins in a dose-dependent manner. CONCLUSIONS: Our study demonstrates that pExo has multiple potential therapeutic effects including symptom control and disease modifying characteristics. This may make it an attractive candidate for further development as an anti-OA therapeutic.


Asunto(s)
Cartílago Articular , Exosomas , Osteoartritis , Humanos , Ratas , Animales , Exosomas/metabolismo , Cartílago Articular/patología , Osteoartritis/metabolismo , Condrocitos/metabolismo , Antiinflamatorios/uso terapéutico
3.
Bioengineering (Basel) ; 10(7)2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37508856

RESUMEN

Chronic wounds are associated with considerable patient morbidity and present a significant economic burden to the healthcare system. Often, chronic wounds are in a state of persistent inflammation and unable to progress to the next phase of wound healing. Placental-derived biomaterials are recognized for their biocompatibility, biodegradability, angiogenic, anti-inflammatory, antimicrobial, antifibrotic, immunomodulatory, and immune privileged properties. As such, placental-derived biomaterials have been used in wound management for more than a century. Placental-derived scaffolds are composed of extracellular matrix (ECM) that can mimic the native tissue, creating a reparative environment to promote ECM remodeling, cell migration, proliferation, and differentiation. Reliable evidence exists throughout the literature to support the safety and effectiveness of placental-derived biomaterials in wound healing. However, differences in source (i.e., anatomical regions of the placenta), preservation techniques, decellularization status, design, and clinical application have not been fully evaluated. This review provides an overview of wound healing and placental-derived biomaterials, summarizes the clinical results of placental-derived scaffolds in wound healing, and suggests directions for future work.

4.
J Mater Sci Mater Med ; 34(7): 37, 2023 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-37486403

RESUMEN

Differences in scaffold design have the potential to influence cell-scaffold interactions. This study sought to determine whether a tri-layer design influences the cellular function of human tenocytes in vitro. The single-layer decellularized, dehydrated human amniotic membrane (DDHAM) and the tri-layer DDHAM (DDHAM-3L) similarly supported tenocyte function as evidenced by improved cell growth and migration, reduced dedifferentiation, and an attenuated inflammatory response. The tri-layer design provides a mechanically more robust scaffold without altering biological activity.


Asunto(s)
Amnios , Tenocitos , Humanos , Proliferación Celular
5.
Cytotherapy ; 25(12): 1265-1270.e2, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37256239

RESUMEN

The tissue factor (TF/CD142) expressed by mesenchymal stromal cells (MSCs) has been regarded as a safety concern in clinical applications as it may trigger thrombosis when MSCs administered intravenously. Human placental allogenic stromal cells (ASCs) are culture-expanded, undifferentiated MSC-like cells derived from full-term postpartum placenta and possess immunomodulatory and pro-angiogenic activities, however, express TF. Here we performed CRISPR/Cas9-mediated TF gene knock out (TFKO) in ASCs, leading to significantly lower TF expression, activity and thrombotic effects. ASCs' characteristics including expansion, expression of phenotypic markers and secretory profile remained unchanged in edited cells, and their immunomodulatory activities, which are functionally relevant to therapeutic applications, were not affected upon TFKO. Taken together, this study provides a feasible strategy which could improve the clinical safety features of MSC-based cell therapy by CRISRP/Cas9-mediated TF gene knock out.


Asunto(s)
Tromboplastina , Trombosis , Femenino , Embarazo , Humanos , Tromboplastina/genética , Sistemas CRISPR-Cas/genética , Placenta , Células del Estroma
6.
J Biomed Mater Res B Appl Biomater ; 111(3): 684-700, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36370413

RESUMEN

Amniotic membrane (AM) is a naturally derived biomaterial with biological and mechanical properties important to Ophthalmology. The epithelial side of the AM promotes epithelialization, while the stromal side regulates inflammation. However, not all AMs are equal. AMs undergo different processing with resultant changes in cellular content and structure. This study evaluates the effects of sidedness and processing on human corneal epithelial cell (HCEC) activity, the effect of processing on HCEC inflammatory response, and then a case study is presented. Three differently processed, commercially available ocular AMs were selected: (1) Biovance®3L Ocular, a decellularized, dehydrated human AM (DDHAM), (2) AMBIO2®, a dehydrated human AM (DHAM), and (3) AmnioGraft®, a cryopreserved human AM (CHAM). HCECs were seeded onto the AMs and incubated for 1, 4 and 7 days. Cell adhesion and viability were evaluated using alamarBlue assay. HCEC migration was evaluated using a scratch wound assay. An inflammatory response was induced by TNF-α treatment. The effect of AM on the expression of pro-inflammatory genes in HCECs was compared using quantitative polymerase chain reaction (qPCR). Staining confirmed complete decellularization and the absence of nuclei in DDHAM. HCEC activity was best supported on the stromal side of DDHAM. Under inflammatory stimulation, DDHAM promoted a higher initial inflammatory response with a declining trend across time. Clinically, DDHAM was used to successfully treat anterior basement membrane dystrophy. Compared with DHAM and CHAM, DDHAM had significant positive effects on the cellular activities of HCECs in vitro, which may suggest greater ocular cell compatibility in vivo.


Asunto(s)
Amnios , Ojo , Humanos , Amnios/metabolismo , Adhesión Celular , Células Epiteliales , Inflamación
7.
Front Immunol ; 13: 900624, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36341337

RESUMEN

Influenza A virus (IAV) infections are a significant recurrent threat to public health and a significant burden on global economy, highlighting the need for developing more effective therapies. Natural killer (NK) cells play a pivotal role in the control of pulmonary IAV infection, however, little is known about the therapeutic potential of adoptively transferred NK cells for viral infections. Here, we investigated the antiviral activity of CYNK, human placental hematopoietic stem cell-derived NK cells, against IAV infection in vitro. Virus infection induced the expression of NK cell activating ligands on respiratory epithelial cells, resulting in enhanced recognition by CYNK cells. Upon co-culture with IAV-infected epithelial cells, CYNK exhibited elevated degranulation and increased production of IFN-γ, TNF-α and GM-CSF in a virus dose-dependent manner. Furthermore, CYNK showed virus dose-dependent cytotoxicity against IAV-infected cells. The antiviral activity of CYNK was mediated by NKp46 and NKG2D. Together, these data demonstrate that CYNK possesses potent antiviral function against IAV and warrant clinical investigations for adoptive NK cell therapies against viral infections.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Embarazo , Humanos , Femenino , Placenta , Células Asesinas Naturales/metabolismo , Gripe Humana/metabolismo , Células Madre Hematopoyéticas , Antivirales/metabolismo
8.
J Exp Orthop ; 9(1): 69, 2022 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-35849201

RESUMEN

PURPOSE: Injectable connective tissue matrices (CTMs) may promote tendon healing, given their minimally invasive properties, structural and biochemical extracellular matrix components, and capacity to fill irregular spaces. The purpose of this study is to evaluate the effects of placental CTMs on the cellular activities of human tenocytes. Decellularization, the removal of cells, cell fragments, and DNA from CTMs, has been shown to reduce the host's inflammatory response. Therefore, the authors hypothesize that a decellularized CTM will provide a more cell-friendly matrix to support tenocyte functions. METHODS: Three human placental CTMs were selected for comparison: AmnioFill® (A-CTM), a minimally manipulated, non-viable cellular particulate, BioRenew™ (B-CTM), a liquid matrix, and Interfyl® (I-CTM), a decellularized flowable particulate. Adhesion and proliferation were evaluated using cell viability assays and tenocyte migration using a transwell migration assay. Gene expression of tenocyte markers, cytokines, growth factors, and matrix metalloprotease (MMP) in tenocytes were assessed using quantitative polymerase chain reaction. RESULTS: Although A-CTM supported more tenocyte adhesion, I-CTM promoted significantly more tenocyte proliferation compared with A-CTM and B-CTM. Unlike A-CTM, tenocyte migration was higher in I-CTM than the control. The presence of I-CTM also prevented the loss of tenocyte phenotype, attenuated the expression of pro-inflammatory cytokines, growth factors, and MMP, and promoted the expression of antifibrotic growth factor, TGFß3. CONCLUSION: Compared with A-CTM and B-CTM, I-CTM interacted more favorably with human tenocytes in vitro. I-CTM supported tenocyte proliferation with reduced de-differentiation and attenuation of the inflammatory response, suggesting that I-CTM may support tendon healing and regeneration in vivo.

9.
Hum Vaccin Immunother ; 18(5): 2055945, 2022 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-35404743

RESUMEN

Influenza A virus (IAV) infections are associated with a high healthcare burden around the world and there is an urgent need to develop more effective therapies. Natural killer (NK) cells have been shown to play a pivotal role in reducing IAV-induced pulmonary infections in preclinical models; however, little is known about the therapeutic potential of adoptively transferred NK cells for IAV infections. Here, we investigated the effects of CYNK-001, human placental hematopoietic stem cell derived NK cells that exhibited strong cytolytic activity against a range of malignant cells and expressed high levels of activating receptors, against IAV infections. In a severe IAV-induced acute lung injury model, mice treated with CYNK-001 showed a milder body weight loss and clinical symptoms, which led to a delayed onset of mortality, thus demonstrating their antiviral protection in vivo. Analysis of bronchoalveolar lavage fluid (BALF) revealed that CYNK-001 reduced proinflammatory cytokines and chemokines highlighting CYNK-001's anti-inflammatory actions in viral induced-lung injury. Furthermore, CYNK-001-treated mice had altered immune responses to IAV with reduced number of neutrophils in BALF yet increased number of CD8+ T cells in the BALF and lung compared to vehicle-treated mice. Our results demonstrate that CYNK-001 displays protective functions against IAV via its anti-inflammatory and immunomodulating activities, which leads to alleviation of disease burden and progression in a severe IAV-infected mice model. The potential of adoptive NK therapy for IAV infections warrants clinical investigation.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Infecciones por Orthomyxoviridae , Animales , Femenino , Células Madre Hematopoyéticas , Humanos , Células Asesinas Naturales , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/prevención & control , Placenta , Embarazo
10.
J Alzheimers Dis ; 86(1): 21-42, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35034899

RESUMEN

The COVID-19 pandemic has accelerated neurological, mental health disorders, and neurocognitive issues. However, there is a lack of inexpensive and efficient brain evaluation and screening systems. As a result, a considerable fraction of patients with neurocognitive or psychobehavioral predicaments either do not get timely diagnosed or fail to receive personalized treatment plans. This is especially true in the elderly populations, wherein only 16% of seniors say they receive regular cognitive evaluations. Therefore, there is a great need for development of an optimized clinical brain screening workflow methodology like what is already in existence for prostate and breast exams. Such a methodology should be designed to facilitate objective early detection and cost-effective treatment of such disorders. In this paper we have reviewed the existing clinical protocols, recent technological advances and suggested reliable clinical workflows for brain screening. Such protocols range from questionnaires and smartphone apps to multi-modality brain mapping and advanced imaging where applicable. To that end, the Society for Brain Mapping and Therapeutics (SBMT) proposes the Brain, Spine and Mental Health Screening (NEUROSCREEN) as a multi-faceted approach. Beside other assessment tools, NEUROSCREEN employs smartphone guided cognitive assessments and quantitative electroencephalography (qEEG) as well as potential genetic testing for cognitive decline risk as inexpensive and effective screening tools to facilitate objective diagnosis, monitor disease progression, and guide personalized treatment interventions. Operationalizing NEUROSCREEN is expected to result in reduced healthcare costs and improving quality of life at national and later, global scales.


Asunto(s)
COVID-19 , Pandemias , Anciano , Encéfalo/diagnóstico por imagen , Mapeo Encefálico , Atención a la Salud , Humanos , Masculino , Calidad de Vida
11.
J Peripher Nerv Syst ; 26(3): 276-289, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34169613

RESUMEN

Neuropathy is a major cause of morbidity and mortality in individuals with diabetes, with no effective therapy to alter the inevitable progression of nerve damage. We hypothesized that mesenchymal stroma cell-like populations, that are characterized as immune modulators also have the potential of inducing angiogenesis and neurite outgrowth, might be useful in treating diabetic peripheral neuropathy (DPN). The aims of this study were to investigate the efficacy and safety of mesenchymal stem cell-like product (PDA-002) in treating DPN. A phase-2 randomized placebo-controlled trial was conducted in 26 patients with DPN. Treatment consisted of three rounds of intramuscular injections in one lower limb using one of the three randomized treatment arms PDA-002 (low-dose 3 × 106 cells), PDA-002 (high-dose 30 × 106 cells), or placebo. Three treatments per patient occurred on days 1, 29, and 57. Study endpoints included efficacy and safety of PDA-002 in treating DPN in both lower extremities following unilateral local injection. Outcome measures included intra-epidermal nerve fiber density (IENFD) up to 1 year from the day of treatment with 6-month as the primary outcome measurement. In this phase 2 study of DPN, PDA-002 was well tolerated in both doses. No significant changes were noted in IENFD in both the treated and untreated leg in the NIS-LL, NTSS-6, or UENS. Mesenchymal stem cells represent a novel mechanism for treating diabetic neuropathy and are well tolerated. Preliminary results highlight the need of further investigation of PDA-001 as a disease modifying agent for treatment of DPN.


Asunto(s)
Neuropatías Diabéticas , Humanos , Diabetes Mellitus Tipo 2 , Neuropatías Diabéticas/tratamiento farmacológico , Proyectos de Investigación
12.
J Immunother Cancer ; 9(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33741730

RESUMEN

BACKGROUND: Tumors often develop resistance to surveillance by endogenous immune cells, which include natural killer (NK) cells. Ex vivo activated and/or expanded NK cells demonstrate cytotoxicity against various tumor cells and are promising therapeutics for adoptive cancer immunotherapy. Genetic modification can further enhance NK effector cell activity or activation sensitization. Here, we evaluated the effect of the genetic deletion of ubiquitin ligase Casitas B-lineage lymphoma pro-oncogene-b (CBLB), a negative regulator of lymphocyte activity, on placental CD34+ cell-derived NK (PNK) cell cytotoxicity against tumor cells. METHODS: Using CRISPR/Cas9 technology, CBLB was knocked out in placenta-derived CD34+ hematopoietic stem cells, followed by differentiation into PNK cells. Cell expansion, phenotype and cytotoxicity against tumor cells were characterized in vitro. The antitumor efficacy of CBLB knockout (KO) PNK cells was tested in an acute myeloid leukemia (HL-60) tumor model in NOD-scid IL2R gammanull (NSG) mice. PNK cell persistence, biodistribution, proliferation, phenotype and antitumor activity were evaluated. RESULTS: 94% of CBLB KO efficacy was achieved using CRISPR/Cas9 gene editing technology. CBLB KO placental CD34+ cells differentiated into PNK cells with high cell yield and >90% purity determined by CD56+ CD3- cell identity. Ablation of CBLB did not impact cell proliferation, NK cell differentiation or phenotypical characteristics of PNK cells. When compared with the unmodified PNK control, CBLB KO PNK cells exhibited higher cytotoxicity against a range of liquid and solid tumor cell lines in vitro. On infusion into busulfan-conditioned NSG mice, CBLB KO PNK cells showed in vivo proliferation and maturation as evidenced by increased expression of CD16, killer Ig-like receptors and NKG2A over 3 weeks. Additionally, CBLB KO PNK cells showed greater antitumor activity in a disseminated HL60-luciferase mouse model compared with unmodified PNK cells. CONCLUSION: CBLB ablation increased PNK cell effector function and proliferative capacity compared with non-modified PNK cells. These data suggest that targeting CBLB may offer therapeutic advantages via enhancing antitumor activities of NK cell therapies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas , Citotoxicidad Inmunológica , Inmunoterapia Adoptiva , Células Asesinas Naturales/trasplante , Neoplasias/terapia , Proteínas Proto-Oncogénicas c-cbl/deficiencia , Células Madre , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antígenos CD34/metabolismo , Proteína 9 Asociada a CRISPR/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Técnicas de Cocultivo , Femenino , Proteínas Ligadas a GPI/metabolismo , Técnicas de Inactivación de Genes , Células HL-60 , Humanos , Células K562 , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Fenotipo , Placenta/citología , Embarazo , Proteínas Proto-Oncogénicas c-cbl/genética , Receptores de IgG/metabolismo , Células Madre/inmunología , Células Madre/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Alzheimers Dis ; 77(2): 459-504, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32925078

RESUMEN

COVID-19 is a severe infectious disease that has claimed >150,000 lives and infected millions in the United States thus far, especially the elderly population. Emerging evidence has shown the virus to cause hemorrhagic and immunologic responses, which impact all organs, including lungs, kidneys, and the brain, as well as extremities. SARS-CoV-2 also affects patients', families', and society's mental health at large. There is growing evidence of re-infection in some patients. The goal of this paper is to provide a comprehensive review of SARS-CoV-2-induced disease, its mechanism of infection, diagnostics, therapeutics, and treatment strategies, while also focusing on less attended aspects by previous studies, including nutritional support, psychological, and rehabilitation of the pandemic and its management. We performed a systematic review of >1,000 articles and included 425 references from online databases, including, PubMed, Google Scholar, and California Baptist University's library. COVID-19 patients go through acute respiratory distress syndrome, cytokine storm, acute hypercoagulable state, and autonomic dysfunction, which must be managed by a multidisciplinary team including nursing, nutrition, and rehabilitation. The elderly population and those who are suffering from Alzheimer's disease and dementia related illnesses seem to be at the higher risk. There are 28 vaccines under development, and new treatment strategies/protocols are being investigated. The future management for COVID-19 should include B-cell and T-cell immunotherapy in combination with emerging prophylaxis. The mental health and illness aspect of COVID-19 are among the most important side effects of this pandemic which requires a national plan for prevention, diagnosis and treatment.


Asunto(s)
Infecciones por Coronavirus , Pandemias , Neumonía Viral , COVID-19 , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/psicología , Infecciones por Coronavirus/terapia , Humanos , Inmunoterapia , Salud Mental , Apoyo Nutricional , Neumonía Viral/diagnóstico , Neumonía Viral/epidemiología , Neumonía Viral/psicología , Neumonía Viral/terapia , Tratamiento Farmacológico de COVID-19
14.
Stem Cells ; 35(6): 1603-1613, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28233380

RESUMEN

Peripheral arterial disease (PAD) is a leading cause of limb loss and mortality worldwide with limited treatment options. Mesenchymal stromal cell (MSC) therapy has demonstrated positive effects on angiogenesis in preclinical models and promising therapeutic efficacy signals in early stage clinical studies; however, the mechanisms underlying MSC-mediated angiogenesis remain largely undefined. Here, we investigated the mechanism of action of human placenta-derived MSC-like cells (PDA-002) in inducing angiogenesis using mice hind limb ischemia model. We showed that PDA-002 improved blood flow and promoted collateral vessel formation in the injured limb. Histological analysis demonstrated that PDA-002 increased M2-like macrophages in ischemic tissue. Analysis of the changes in functional T cell phenotype in the draining lymph nodes revealed that PDA-002 treatment was associated with the induction of cytokine and gene expression signatures of Th2 response. Angiogenic effect of PDA-002 was markedly reduced in Balb/c nude mice compared with wild type. This reduction in efficacy was reversed by T cell reconstitution, suggesting T cells are essential for PDA-002-mediated angiogenesis. Furthermore, effect of PDA-002 on macrophage differentiation was also T cell-dependent as a PDA-002-mediated M2-like macrophage skewing was only observed in wild type and T cell reconstituted nude mice, but not in nude mice. Finally, we showed that PDA-002-treated animals had enhanced angiogenic recovery in response to the second injury when PDA-002 no longer persisted in vivo. These results suggest that PDA-002 enhances angiogenesis through an immunomodulatory mechanism involving T cell-dependent reprogramming of macrophage differentiation toward M2-like phenotype. Stem Cells 2017;35:1603-1613.


Asunto(s)
Diferenciación Celular , Macrófagos/citología , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica , Placenta/citología , Linfocitos T/citología , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Isquemia/patología , Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Perfusión , Fenotipo , Embarazo , Linfocitos T/metabolismo
15.
Wounds ; 29(1): 28-38, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27852013

RESUMEN

BACKGROUND: Decellularized, dehydrated human amniotic membrane (DDHAM) is an extracellular matrix devoid of cells, cell debris, and growth factors. This study examines the effect of cell attachment to the DDHAM and the induced cellular responses. MATERIALS AND METHODS: The cell types employed in this study were human dermal fibroblasts (HDF), human epithelial keratinocytes (HEK), and human dermal microvascular endothelial cells (HDMEC), all of which play critical roles in the wound healing process. Further, the DDHAM was compared to a dehydrated human amnion/chorion membrane (dHACM), which contains and releases biological entities including growth factors and cytokines. The HDF and HEK were cultured on the DDHAM and the dHACM, and cell imaging and proliferation assays were performed to evaluate cell attachment to and the ability to proliferate on the DDHAM relative to the dHACM. In addition, the effect of soluble factors released by the DDHAM and the dHACM on cell survival, attachment, and proliferation were examined. The authors also evaluated the effect of soluble factors produced by culturing cells on the DDHAM in in vitro functional assays, including cell survival and endothelial cell migration in a wound closure angiogenesis assay. RESULTS: The HDF and HEK cells readily attached to and proliferated on the DDHAM, while the dHACM did not support cell attachment and proliferation when cultured under the same conditions. Soluble factors secreted when HDF were cultured on the DDHAM enhanced both endothelial cell and keratinocyte survival and endothelial cell migration in a wound closure assay. CONCLUSIONS: Although DDHAM is only an extracellular matrix and serves primarily as a scaffold, it has sufficient cues to allow for cell attachment and proliferation. Further, the biological entities released as a consequence of cell attachment promote cell survival and migration.


Asunto(s)
Amnios/química , Apósitos Biológicos , Adhesión Celular , Proliferación Celular , Neovascularización Fisiológica , Heridas y Lesiones/patología , Aloinjertos , Uniones Célula-Matriz , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestructura , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Técnicas In Vitro , Resultado del Tratamiento , Cicatrización de Heridas
16.
J Vasc Surg ; 64(3): 746-756.e1, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26054585

RESUMEN

OBJECTIVE: Human placenta-derived adherent cells (PDACs) are a culture-expanded, undifferentiated mesenchymal-like population from full-term placental tissue and were previously shown to possess anti-inflammatory and immunomodulatory properties. PDACs (formulated as PDA-002) are in clinical trials for peripheral arterial disease with diabetic foot ulcer. In the current study, we examined their angiogenic and tissue reparative properties. METHODS: The effects of PDACs on survival and tube formation of human umbilical vein endothelial cells (HUVECs) were tested using conditioned media and noncontact coculture. Angiogenic effects were assessed in the chick chorioallantoic membrane assay. Hindlimb ischemia (HLI) was induced in mice and rats by femoral artery transection, and blood flow and blood vessel density were monitored in vivo by laser Doppler and angiography in the ischemic and control limbs. Tissue damage and regeneration in HLI were examined in histologic sections of quadriceps muscle stained with hematoxylin and eosin, and newly synthesized blood vessels were detected by indoxyl-tetrazolium staining for alkaline phosphatase. RESULTS: PDACs enhanced the survival of serum-starved HUVECs and stimulated HUVEC tube formation, and in the chick chorioallantoic membrane assay, PDACs stimulated blood vessel formation. In HLI, intramuscular administration of PDACs resulted in improved blood flow and vascular density, and in quadriceps muscle, tissue regeneration and increased numbers of blood vessels were observed. CONCLUSIONS: PDACs exhibited various activities consistent with angiogenesis and tissue repair, supporting the continued investigation of this cell therapy as treatment for vascular disease-related indications.


Asunto(s)
Adhesión Celular , Membrana Corioalantoides/irrigación sanguínea , Isquemia/cirugía , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Neovascularización Fisiológica , Placenta/citología , Músculo Cuádriceps/irrigación sanguínea , Animales , Velocidad del Flujo Sanguíneo , Células Cultivadas , Embrión de Pollo , Técnicas de Cocultivo , Medios de Cultivo Condicionados/metabolismo , Modelos Animales de Enfermedad , Femenino , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Isquemia/metabolismo , Isquemia/fisiopatología , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos BALB C , Comunicación Paracrina , Embarazo , Ratas Sprague-Dawley , Recuperación de la Función , Flujo Sanguíneo Regional , Factores de Tiempo
17.
Food Funct ; 6(7): 2303-14, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26073176

RESUMEN

Chicken egg yolk is a rich source of nutrients providing high quality proteins, vitamins, minerals, carotenoids and antioxidants. Chicken egg yolk, recovered from whole egg within 24 hours post-lay has been utilized as a starting material in the preparation of a dietary supplement that has been demonstrated to lead to gains in muscle mass in a human clinical study. Further, an oil derived from chicken egg yolk has been utilized as a topical agent to treat third degree burn injury. The molecular changes that take place in fertilized, chicken egg yolk during the first 24 hours post-lay are not well understood. By studying how the protein composition of egg yolk varies with fertility status, one can utilize this knowledge to develop egg yolk-based products that have been optimized for specific applications. In this study, a direct quantitative comparison was made between the proteome of fertilized chicken egg yolk and the proteome of unfertilized chicken egg yolk, both maintained at 20 °C and analyzed within 24 hours post-lay. Egg yolk proteins from each fertility state were digested with trypsin, labeled with distinct chemical labels (tandem mass tag reagents) and then combined in a 1 : 1 ratio. A TMT-labeled tryptic digest derived from chicken egg yolk proteins (fertilized and unfertilized) was separated using high-pH/low-pH reverse-phase chromatography and analyzed using mass spectrometry. 225 protein identifications were made from this TMT-labeled tryptic digest based on a minimum of 2 unique peptides observed per protein. 9 proteins increased in abundance in fertilized egg yolk relative to unfertilized egg yolk and 9 proteins decreased in abundance in fertilized egg yolk relative to unfertilized egg yolk. Some proteins that increased in abundance in fertilized egg yolk play an important role in angiogenesis (pleiotrophin, histidine rich glycoprotein) and defense against pathogens (mannose-binding lectin, ß-defensin 11, serum amyloid P-component, ovostatin). Based on this study, fertilized chicken egg yolk may be more useful as a starting material relative to unfertilized chicken egg yolk for the purpose of enriching or isolating proteins with pro-angiogenic and anti-microbial properties.


Asunto(s)
Proteínas del Huevo/química , Yema de Huevo/química , Proteoma/química , Secuencia de Aminoácidos , Animales , Pollos , Cromatografía de Fase Inversa/instrumentación , Proteínas del Huevo/genética , Proteínas del Huevo/metabolismo , Yema de Huevo/metabolismo , Fertilización , Datos de Secuencia Molecular , Proteoma/genética , Proteoma/metabolismo , Alineación de Secuencia , Espectrometría de Masas en Tándem , Temperatura
18.
Stem Cells Transl Med ; 4(7): 695-701, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26019227

RESUMEN

UNLABELLED: SummaryIt has been assumed that adult tissues cannot regenerate themselves. With the current understanding that every adult tissue has its own intrinsic progenitor or stem cell, it is now clear that almost all tissues have regenerative potential partially related to their innate turnover dynamics. Moreover, it appears that a separate class of local cells originating as perivascular cells appears to provide regulatory oversight for localized tissue regeneration. The management of this regeneration oversight has a profound influence on the use of specific cells for cell therapies as a health care delivery tool set. The multipotent mesenchymal stem cell (MSC), now renamed the medicinal signaling cell, predominantly arises from pericytes released from broken and inflamed blood vessels and appears to function as both an immunomodulatory and a regeneration mediator. MSCs are being tested for their management capabilities to produce therapeutic outcomes in more than 480 clinical trials for a wide range of clinical conditions. Local MSCs function by managing the body's primary repair and regeneration activities. Supplemental MSCs can be provided from either endogenous or exogenous sources of either allogeneic or autologous origin. This MSC-based therapy has the potential to change how health care is delivered. These medicinal cells are capable of sensing their surroundings. Also, by using its complex signaling circuitry, these cells organize site-specific regenerative responses as if these therapeutic cells were well-programmed modern computers. Given these facts, it appears that we are entering a new age of cellular medicine. SIGNIFICANCE: This report is a perspective from an active scientist and an active entrepreneur and commercial leader. It is neither a comprehensive review nor a narrowly focused treatise. The broad themes and the analogy to the working component of a computer and that of a cell are meant to draw several important scientific principles and health care themes together into the thesis that regenerative medicine is a constant throughout life and its management is the next frontier of health care. Mesenchymal stem cells are used as the central connection in the broad theme, not as multipotent progenitors but rather as an important control element in the natural local regeneration process.

19.
Inflamm Bowel Dis ; 21(8): 1809-16, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25985246

RESUMEN

BACKGROUND: PDA-001 (cenplacel-L), a preparation of placenta-derived mesenchymal-like adherent cells with immunomodulatory effects, previously demonstrated safety and tolerability in an open-label Crohn's disease (CD) study. The current phase 1b/2a study evaluated the safety and efficacy of PDA-001 in subjects with moderate-to-severe CD. METHODS: Subjects had active inflammation on colonoscopy or elevated fecal calprotectin and inadequate response to conventional therapy. Concomitant therapy with stable doses of immunomodulators and/or biologics was permitted. Subjects received 8 units of PDA-001 (1.5 × 10 cells per unit) in the phase 1b open-label study. In the phase 2a double-blind study, subjects were randomly assigned placebo, 1 unit, or 4 units of PDA-001 (2 infusions 1 wk apart). The primary endpoint was induction of clinical response (≥100 points and/or 25% decrease in Crohn's Disease Activity Index) at 4 and 6 weeks. RESULTS: Fifty subjects were enrolled (safety analysis, 50 subjects; efficacy analysis, 48 subjects). Four subjects received 8 units of PDA-001 (phase 1b study); 46 subjects were subsequently randomized to 1 or 4 units of PDA-001 or placebo (phase 2a study). The primary endpoint was achieved in 10/28 (36%) of PDA-001 subjects compared with placebo (0%, P = 0.026). Clinical remission was achieved in 4/28 (14%) of PDA-001 subjects compared with placebo (0%, P = 0.3). One treatment-related serious adverse event occurred (systemic hypersensitivity reaction at 8 units). In the phase 2a study, serious adverse events occurred in 9/28 (32%) of PDA-001 subjects and 1/16 (7%) of placebo subjects. CONCLUSIONS: A 2-infusion regimen of PDA-001 induced clinical response in subjects with moderate-to-severe CD. Additional studies are warranted.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Enfermedad de Crohn/patología , Enfermedad de Crohn/terapia , Placenta/citología , Adolescente , Adulto , Anciano , Células Cultivadas , Método Doble Ciego , Femenino , Estudios de Seguimiento , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Embarazo , Pronóstico , Calidad de Vida , Inducción de Remisión , Seguridad , Adulto Joven
20.
Stem Cells Transl Med ; 4(3): 269-75, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25673767

RESUMEN

Human placenta-derived adherent cells (PDACs) are a culture-expanded, undifferentiated mesenchymal-like population derived from full-term placental tissue, with immunomodulatory, anti-inflammatory, angiogenic, and neuroprotective properties. PDA-001 (cenplacel-L), an intravenous formulation of PDAC cells, is in clinical development for the treatment of autoimmune and inflammatory diseases. We tested the therapeutic effects of PDA-001 in mice with chronic heart failure (CHF). Three weeks after transaortic constriction surgery to induce CHF, the mice underwent direct intramyocardial (IM) or i.v. injection of PDA-001 at a high (0.5 × 10(6) cells per mouse), medium (0.5 × 10(5) cells per mouse), or low (0.5 × 10(4) cells per mouse) dose. The mice were sacrificed 4 weeks after treatment. Echocardiography and ventricular catheterization showed that IM injection of PDA-001 significantly improved left ventricular systolic and diastolic function compared with injection of vehicle or i.v. injection of PDA-001. IM injection of PDA-001 also decreased cardiac fibrosis, shown by trichrome staining in the vicinity of the injection sites. Low-dose treatment showed the best improvement in cardiac performance compared with the medium- and high-dose groups. In another independent study to determine the mechanism of action with bromodeoxyuridine labeling, the proliferation rates of endothelial cells and cardiomyocytes were significantly increased by low or medium IM dose PDA-001. However, no surviving PDA-001 cells were detected in the heart 1 month after injection. In vivo real-time imaging consistently revealed that the PDA-001 cells were detectable only within 2 days after IM injection of luciferase-expressing PDA-001. Together, these results have demonstrated the cardiac therapeutic potential of PDA-001, likely through a paracrine effect.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Insuficiencia Cardíaca/terapia , Placenta , Animales , Adhesión Celular , Enfermedad Crónica , Femenino , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Xenoinjertos , Humanos , Ratones , Miocitos Cardíacos/metabolismo , Placenta/citología , Placenta/metabolismo , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...