Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Regen Med ; 19(1): 47-63, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38240144

RESUMEN

Aim: Cell therapies for diabetes rely on differentiation of stem cells into insulin-producing cells, which is complex and expensive. Our goal was to evaluate production costs and test ways to reduce it. Methods: Cost of Goods (COGs) analysis for differentiation was completed and the effects of replacement or reduction of the most expensive item was tested using qRT-PCR, immunohistochemistry, flow cytometry along with glucose-stimulated insulin release. Results: Activin A (AA) was responsible for significant cost. Replacement with small molecules failed to form definitive endoderm (DE). Reducing AA by 50% did not negatively affect expression of beta cell markers. Conclusion: Reduction of AA concentration is feasible without adversely affecting DE and islet-like cell differentiation, leading to significant cost savings in manufacturing.


Asunto(s)
Endodermo , Insulinas , Endodermo/metabolismo , Diferenciación Celular , Activinas/metabolismo , Activinas/farmacología , Insulinas/metabolismo
2.
PLoS One ; 17(5): e0267814, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35613086

RESUMEN

BACKGROUND: Protection of islets without systemic immunosuppression has been a long-sought goal in the islet transplant field. We conducted a pilot biocompatibility/safety study in healthy dogs followed by a dose-finding efficacy study in diabetic dogs using polyethylene glycol diacrylate (PEGDA) microencapsulated allogeneic canine islets. METHODS: Prior to the transplants, characterization of the canine islets included the calculations determining the average cell number/islet equivalent. Following measurements of purity, insulin secretion, and insulin, DNA and ATP content, the islets were encapsulated and transplanted interperitoneally into dogs via a catheter, which predominantly attached to the omentum. In the healthy dogs, half of the microspheres injected contained canine islets, the other half of the omentum received empty PEGDA microspheres. RESULTS: In the biocompatibility study, healthy dogs received increasing doses of cells up to 1.7 M cells/kg body weight, yet no hypoglycemic events were recorded and the dogs presented with no adverse events. At necropsy the microspheres were identified and described as clear with attachment to the omentum. Several of the blood chemistry values that were abnormal prior to the transplants normalized after the transplant. The same observation was made for the diabetic dogs that received higher doses of canine islets. In all diabetic dogs, the insulin required to attempt to control blood glucose was cut by 50-100% after the transplant, down to no required insulin for the course of the 60-day study. The dogs had no adverse events and behavioral monitoring suggested normal activity after recovery from the transplant. CONCLUSIONS AND IMPLICATIONS: The study provides evidence that PEGDA microencapsulated canine islets reversed the signs of diabetes without immunosuppression and led to states of insulin-independence or significantly lowered insulin requirements in the recipients.


Asunto(s)
Diabetes Mellitus , Trasplante de Células Madre Hematopoyéticas , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Animales , Glucemia , Diabetes Mellitus/terapia , Diabetes Mellitus/veterinaria , Perros , Terapia de Inmunosupresión , Insulina , Polietilenglicoles
3.
BMC Vet Res ; 17(1): 244, 2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-34266445

RESUMEN

BACKGROUND: The source of multipotent stromal cells (MSC) can have a significant influence on the health and expansion capacity of the cells. As the applications for allogeneic MSCs in the treatment of feline diseases increase, the location of the initial donor tissue must be analyzed. To date, comparisons have only been made between feline MSCs collected from bone marrow or abdominal fat. This is the first report to compare cells obtained from different adipose depots in the cat with a focus on clinically relevant donor tissues. The tissue was collected from 34 healthy cats undergoing spaying (fat around the ovaries and uterine horn) or subcutaneous fat collected during surgical procedures. RESULTS: The amount of starting material is essential to isolate sufficient MSCs. The total tissue yield from the subcutaneous fat was significantly greater than could be obtained from around the reproductive organs, leading to 3 times more MSCs per donor. However, the concentration of MSCs obtained from reproductive fat was higher than from subcutaneous fat. In addition, the viability of the MSCs from the reproductive fat was significantly higher than the subcutaneous fat. Since most spaying occurs in young cats (under 18 months) reproductive fat was collected from adult cats during spaying, illustrating that age did not alter the yield or viability of the MSCs. When sufficient tissue was collected, it was digested either mechanically or enzymatically. Mechanical digestion further decreased the viability and yield of MSCs from subcutaneous fat compared to enzymatic digestion. Biomarkers of stem cell characterization, expansion capacity and function were detected using qPCR. CD70, CD90 and CD105 were all expressed in high levels in the 3 groups. However, the reproductive fat had higher levels of CD73 with the mechanically digested subcutaneous fat having the least. Gata6 was detected in all samples while Sox2 and Sox17 were also detected with higher quantities found in the enzymatically digested subcutaneous fat. Negative control genes of Gata4 and Pdx1 showed no detection prior to 50 cycles. During the first three passages, age of the donor, location of the donor tissue, or digestion protocol had no effect on cell culture doubling times or cell viability. CONCLUSIONS: While MSCs from reproductive fat had superior cells/tissue weight and initial viability, there were still dramatically fewer cells obtained compared to subcutaneous fat due to the limited amount of tissue surrounding the reproductive organs. Further, in P1-P3 cultures there were no differences noted in doubling time or cell viability between tissue obtained from reproductive or subcutaneous fat depots.


Asunto(s)
Gatos , Grasa Intraabdominal/citología , Células Madre Mesenquimatosas/citología , Grasa Subcutánea/citología , Animales , Técnicas de Cultivo de Célula/métodos , Técnicas de Cultivo de Célula/veterinaria , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Femenino , Genitales Femeninos/cirugía , Masculino , Células Madre Mesenquimatosas/fisiología
4.
Tissue Eng Part A ; 27(3-4): 153-164, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32103710

RESUMEN

Cell microencapsulation is a rapidly expanding field with broad potential for stem cell therapies and tissue engineering research. Traditional alginate microspheres suffer from poor biocompatibility, and microencapsulation of more advanced hydrogels is challenging due to their slower gelation rates. We have developed a novel, noncytotoxic, nonemulsion-based method to produce hydrogel microspheres compatible with a wide variety of materials, called core-shell spherification (CSS). Fabrication of microspheres by CSS derived from two slow-hardening hydrogels, hyaluronic acid (HA) and polyethylene glycol diacrylate (PEGDA), was characterized. HA microspheres were manufactured with two different crosslinking methods: thiolation and methacrylation. Microspheres of methacrylated HA (MeHA) had the greatest swelling ratio, the largest average diameter, and the lowest diffusion barrier. In contrast, PEGDA microspheres had the smallest diameters, the lowest swelling ratio, and the highest diffusion barrier, while microspheres of thiolated HA had characteristics that were in between the other two groups. To test the ability of the hydrogels to protect cells, while promoting function, diabetic NOD mice received intraperitoneal injections of PEGDA or MeHA microencapsulated canine islets. PEGDA microspheres reversed diabetes for the length of the study (up to 16 weeks). In contrast, islets encapsulated in MeHA microspheres at the same dose restored normoglycemia, but only transiently (3-4 weeks). Nonencapsulated canine islet transplanted at the same dose did not restore normoglycemia for any length of time. In conclusion, CSS provides a nontoxic microencapsulation procedure compatible with various hydrogel types.


Asunto(s)
Ácido Hialurónico , Polietilenglicoles , Alginatos , Animales , Perros , Hidrogeles , Ratones , Ratones Endogámicos NOD , Microesferas
5.
Regen Med ; 15(5): 1661-1678, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32589107

RESUMEN

The field of cell therapy has blossomed, providing exciting new options for treating a variety of diseases. While few cell therapy products have US FDA approval, there are thousands of cell treatments at various stages of development, pointing to a potential revolutionary shift in patient care. The expanding number and nature of cellular therapies necessitate greater standardization. Several international organizations are collaborating to pursue some level of global standardization, especially concerning cell banking. However, less harmonization surrounds assays used for critical quality characterization including: identity, purity, safety and potency. Frequently, there is divergence regarding the terms describing the characterization assays across regulatory authorities and guidances. This review summarizes the critical quality assays currently used for different categories of cell therapies. Areas of harmonization and an absence of standardization are highlighted. We propose potential solutions to facilitate harmonization of critical quality characterization assays and the language used to describe them.


Asunto(s)
Bioensayo/métodos , Bioensayo/normas , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/normas , Animales , Humanos , Control de Calidad
6.
Clin Transl Med ; 5(Suppl 1): 26, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27558513

RESUMEN

TABLE OF CONTENTS: A1 One health advances and successes in comparative medicine and translational researchCheryl StroudA2 Dendritic cell-targeted gorilla adenoviral vector for cancer vaccination for canine melanomaIgor Dmitriev, Elena Kashentseva, Jeffrey N. Bryan, David T. CurielA3 Viroimmunotherapy for malignant melanoma in the companion dog modelJeffrey N. Bryan, David Curiel, Igor Dmitriev, Elena Kashentseva, Hans Rindt, Carol Reinero, Carolyn J. HenryA4 Of mice and men (and dogs!): development of a commercially licensed xenogeneic DNA vaccine for companion animals with malignant melanomaPhilip J. BergmanA5 Successful immunotherapy with a recombinant HER2-expressing Listeria monocytogenes in dogs with spontaneous osteosarcoma paves the way for advances in pediatric osteosarcomaNicola J. Mason, Josephine S. Gnanandarajah, Julie B. Engiles, Falon Gray, Danielle Laughlin, Anita Gaurnier-Hausser, Anu Wallecha, Margie Huebner, Yvonne PatersonA6 Human clinical development of ADXS-HER2Daniel O'ConnorA7 Leveraging use of data for both human and veterinary benefitLaura S. TremlA8 Biologic replacement of the knee: innovations and early clinical resultsJames P. StannardA9 Mizzou BioJoint Center: a translational success storyJames L. CookA10 University and industry translational partnership: from the lab to commercializationMarc JacobsA11 Beyond docking: an evolutionarily guided OneHealth approach to drug discoveryGerald J. Wyckoff, Lee Likins, Ubadah Sabbagh, Andrew SkaffA12 Challenges and opportunities for data applications in animal health: from precision medicine to precision husbandryAmado S. GuloyA13 A cloud-based programmable platform for healthHarlen D. HaysA14 Comparative oncology: One Health in actionAmy K. LeBlancA15 Companion animal diseases bridge the translational gap for human neurodegenerative diseaseJoan R. Coates, Martin L. Katz, Leslie A. Lyons, Gayle C. Johnson, Gary S. Johnson, Dennis P. O'BrienA16 Duchenne muscular dystrophy gene therapyDongsheng DuanA17 Polycystic kidney disease: cellular mechanisms to emerging therapiesJames P. CalvetA18 The domestic cat as a large animal model for polycystic kidney diseaseLeslie A. Lyons, Barbara GandolfiA19 The support of basic and clinical research by the Polycystic Kidney Disease FoundationDavid A. BaronA20 Using naturally occurring large animal models of human disease to enable clinical translation: treatment of arthritis using autologous stromal vascular fraction in dogsMark L. WeissA21 Regulatory requirements regarding clinical use of human cells, tissues, and tissue-based productsDebra A. WebsterA22 Regenerative medicine approaches to Type 1 diabetes treatmentFrancis N. KaranuA23 The zoobiquity of canine diabetes mellitus, man's best friend is a friend indeed-islet transplantationEdward J. RobbA24 One Medicine: a development model for cellular therapy of diabetesRobert J. Harman.

7.
Cell Stem Cell ; 13(2): 175-89, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23910084

RESUMEN

Numerous studies have shown that the bone marrow (BM) niche plays a key role in mouse hematopoietic stem cell (HSC) function and involves contributions from a broad array of cell types. However, the composition and role of the human BM HSC niche have not been investigated. Here, using human bone biopsy specimens, we provide evidence of HSC propensity to localize to endosteal regions of the trabecular bone area (TBA). Through functional xenograft transplantation, we found that human HSCs localizing to the TBA have superior regenerative and self-renewal capacity and are molecularly distinct from those localizing to the long bone area (LBA). In addition, osteoblasts in the TBA possess unique characteristics and express a key network of factors that regulate TBA- versus LBA-localized human HSCs in vivo. Our study reveals that BM localization and architecture play a critical role in defining the functional and molecular properties of human HSCs.


Asunto(s)
Células de la Médula Ósea/metabolismo , Huesos/patología , Células Madre Hematopoyéticas/metabolismo , Animales , Biopsia , Células de la Médula Ósea/patología , Proliferación Celular , Células Madre Hematopoyéticas/patología , Humanos , Ligandos , Ratones , Osteoblastos/metabolismo , Osteoblastos/patología , Receptores Notch/metabolismo , Nicho de Células Madre , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Diabetes ; 61(8): 2016-29, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22740171

RESUMEN

Diabetes is a chronic debilitating disease that results from insufficient production of insulin from pancreatic ß-cells. Islet cell replacement can effectively treat diabetes but is currently severely limited by the reliance upon cadaveric donor tissue. We have developed a protocol to efficiently differentiate commercially available human embryonic stem cells (hESCs) in vitro into a highly enriched PDX1+ pancreatic progenitor cell population that further develops in vivo to mature pancreatic endocrine cells. Immature pancreatic precursor cells were transplanted into immunodeficient mice with streptozotocin-induced diabetes, and glycemia was initially controlled with exogenous insulin. As graft-derived insulin levels increased over time, diabetic mice were weaned from exogenous insulin and human C-peptide secretion was eventually regulated by meal and glucose challenges. Similar differentiation of pancreatic precursor cells was observed after transplant in immunodeficient rats. Throughout the in vivo maturation period hESC-derived endocrine cells exhibited gene and protein expression profiles that were remarkably similar to the developing human fetal pancreas. Our findings support the feasibility of using differentiated hESCs as an alternative to cadaveric islets for treating patients with diabetes.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Células Secretoras de Insulina/citología , Páncreas/citología , Animales , Línea Celular , Diabetes Mellitus Experimental/terapia , Proteínas de Homeodominio/biosíntesis , Humanos , Insulina/uso terapéutico , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Páncreas/embriología , Proproteína Convertasas/biosíntesis , Ratas , Células Madre/citología , Transactivadores/biosíntesis
9.
Diabetes ; 60(1): 239-47, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20971966

RESUMEN

OBJECTIVE: Differentiation of human embryonic stem (hES) cells to fully developed cell types holds great therapeutic promise. Despite significant progress, the conversion of hES cells to stable, fully differentiated endocrine cells that exhibit physiologically regulated hormone secretion has not yet been achieved. Here we describe an efficient differentiation protocol for the in vitro conversion of hES cells to functional glucagon-producing α- cells. RESEARCH DESIGN AND METHODS: Using a combination of small molecule screening and empirical testing, we developed a six-stage differentiation protocol for creating functional α-cells. An extensive in vitro and in vivo characterization of the differentiated cells was performed. RESULTS: A high rate of synaptophysin expression (>75%) and robust expression of glucagon and the α-cell transcription factor ARX was achieved. After a transient polyhormonal state in which cells coexpress glucagon and insulin, maturation in vitro or in vivo resulted in depletion of insulin and other ß-cell markers with concomitant enrichment of α-cell markers. After transplantation, these cells secreted fully processed, biologically active glucagon in response to physiologic stimuli including prolonged fasting and amino acid challenge. Moreover, glucagon release from transplanted cells was sufficient to reduce demand for pancreatic glucagon, resulting in a significant decrease in pancreatic α-cell mass. CONCLUSIONS: These results indicate that fully differentiated pancreatic endocrine cells can be created via stepwise differentiation of hES cells. These cells may serve as a useful screening tool for the identification of compounds that modulate glucagon secretion as well as those that promote the transdifferentiation of α-cells to ß-cells.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Células Secretoras de Glucagón/citología , Glucagón/metabolismo , Animales , Glucemia/metabolismo , Línea Celular , ADN/análisis , ADN/genética , Células Madre Embrionarias/metabolismo , Glucagón/análisis , Células Secretoras de Glucagón/metabolismo , Humanos , Inmunohistoquímica , Insulina/análisis , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Ratones , Proglucagón/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Differentiation ; 80(2-3): 130-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20561745

RESUMEN

Islet transplantation represents a potential cure for type 1 diabetes; however, a lack of sufficient donor material limits its clinical use. To address the shortfall of islet availability, surrogate insulin-producing cells are sought. Studies suggest that human amniotic fluid (hAF) contains multipotent progenitor cells capable of differentiating to all three germ layers. Here, we used high-content, live-cell imaging to assess the ability to reprogram hAF cells towards a beta cell phenotype. A fluorescent reporter system was developed where DsRed express (DSRE) expression is driven by the human insulin promoter. Using integrative lentiviral technology, we created stable reporter hAF cells that could be routinely monitored for insulin promoter activation. These cells were subjected to combinatorial high-content screening using adenoviral-mediated expression of up to six transcription factors important for beta cell development. Cells were monitored for DSRE expression which revealed an optimal combination of the transcription factors required to induce insulin gene expression in hAF cells. These optimally induced cells were examined for expression of additional beta cell transcription factors and proteins involved in glucose sensing and insulin processing. RT-qPCR revealed very low level expression of insulin that was ultimately insufficient to reverse streptozotocin-induced diabetes following sub-capsular kidney transplantation. High-content, live-cell imaging using fluorescent reporter cells provides a convenient method for repeated assessment of cellular reprogramming. hAF cells could be reprogrammed to express key beta cell proteins, however insulin gene expression was insufficient to reverse hyperglycemia in diabetic animals.


Asunto(s)
Líquido Amniótico/citología , Insulina/metabolismo , Adenoviridae/genética , Animales , Células Cultivadas , Citometría de Flujo , Genes Reporteros , Humanos , Inmunohistoquímica , Insulina/genética , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Regiones Promotoras Genéticas , Ratas , Factores de Transcripción/metabolismo
11.
Dev Cell ; 8(5): 651-63, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15866157

RESUMEN

The molecular basis governing functional behavior of human hematopoietic stem cells (HSCs) is largely unknown. Here, using in vitro and in vivo assays, we isolate and define progenitors versus repopulating HSCs from multiple stages of human development for global gene expression profiling. Accounting for both the hierarchical relationship between repopulating cells and their progenitors, and the enhanced HSC function unique to early stages of ontogeny, the human homologs of Hairy Enhancer of Split-1 (HES-1) and Hepatocyte Leukemia Factor (HLF) were identified as candidate regulators of HSCs. Transgenic human hematopoietic cells expressing HES-1 or HLF demonstrated enhanced in vivo reconstitution ability that correlated to increased cycling frequency and inhibition of apoptosis, respectively. Our report identifies regulatory factors involved in HSC function that elicit their effect through independent systems, suggesting that a unique orchestration of pathways fundamental to all human cells is capable of controlling stem cell behavior.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Hematopoyesis/fisiología , Trasplante de Células Madre Hematopoyéticas , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factor de Transcripción HES-1 , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Transfección , Trasplante Heterólogo
12.
Haematologica ; 90(2): 166-72, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15710567

RESUMEN

BACKGROUND AND OBJECTIVES: Ex vivo expansion of primitive hematopoietic cells for transplantation is an important step to realizing the optimal clinical potential of human cord blood (CB). We aimed to characterize minimal growth factor (GF) conditions that allow ex vivo expansion of primitive cells, including candidate hematopoietic stem cells. DESIGN AND METHODS: Here, we directly investigated the effect of thrombopoietin (TPO) on progenitors and repopulating cells using serum-free culture of CB Lin-CD34+CD38- cells in two different minimal GF conditions: stem cell factor (SCF)+FLT-3-L (termed S/F) and SCF+FLT-3-L+TPO (termed S/F/T). RESULTS: While S/F media supported only low levels of total cell and CFU (colony-forming unit) expansion, the addition of TPO (S/F/T) partially restored cell proliferation, and completely restored CFU expansion to levels observed using full GF conditions (SCF+FLT-3-L+interleukin (IL)-3 (IL-3)+ IL-6+ granulocyte colony-stimulating factor (G-CSF). Intravenous transplantation of either S/F- or S/F/T-expanded cells into NOD/SCID mice resulted in similar frequencies and levels of multilineage reconstitution. INTERPRETATION AND CONCLUSIONS: The use of minimal cytokine stimulation and simultaneous assessment of CFU and SRC indicate that hematopoietic progenitors and in vivo-detected repopulating cells are differentially responsive to TPO; CFU expand in response to TPO but SRC do not. In addition, our study suggests that TPO can functionally replace IL-3+IL-6+G-CSF for CFU expansion of ex vivo cultured CB Lin-CD34+CD38- hematopoietic stem cells.


Asunto(s)
Técnicas de Cultivo de Célula , Sangre Fetal/citología , Sustancias de Crecimiento/metabolismo , Células Madre/citología , ADP-Ribosil Ciclasa 1/biosíntesis , Animales , Antígenos CD34/biosíntesis , Proliferación Celular , Trasplante de Células , Medio de Cultivo Libre de Suero/metabolismo , Humanos , Interleucina-3/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID
13.
Parasite Immunol ; 25(2): 59-68, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12791101

RESUMEN

Immune responses to Haemonchus contortus intestinal antigens were evaluated using abomasal lymph node (ALN) lymphocytes from kid goats protected against challenge infection by immunization with parasite intestinal antigen, and from kids that were challenged after immunization with ovalbumin. ALN lymphocytes from the intestinal antigen-immunized group produced significantly higher antibody levels against intestinal antigens than the ovalbumin group, supporting the theory that immunization contributed to that ALN response. In contrast, intestinal lysates and membrane enriched preparations from intestinal cells stimulated significant proliferation of ALN lymphocytes in both groups. The proliferation was antigen-dependent, since intestinal antigens failed to stimulate proliferation in ALN lymphocytes from unimmunized and uninfected kids. For both the intestinal antigen and ovalbumin immunized groups, CD4+ T lymphocytes predominated in ALN lymphocytes that were stimulated to proliferate by intestinal antigens. The results indicate that H. contortus infection alone can induce ALN lymphocyte responses to intestinal antigens. In contrast to ALN lymphocyte responses, serum antibody against intestinal antigens was generally low to undetectable in ovalbumin-immunized kids following infection. Abomasal mucus from an H. contortus infected lamb was probed with a monoclonal antibody that binds to a periodate sensitive determinant on numerous H. contortus intestinal membrane and secreted proteins. Numerous bands of reactivity were detected, indicating that multiple parasite intestinal antigens were released into abomasal mucus during infection. The results, challenge the general concept that H. contortus intestinal antigens are 'hidden' from the host immune system during an infection. On the contrary, parasite intestinal proteins may be relatively abundant antigens presented to the host during infection. In addition, ALN T lymphocytes appear to provide a more sensitive measure than serum antibody to detect presentation of these antigens to the host immune system.


Asunto(s)
Abomaso/inmunología , Antígenos Helmínticos/inmunología , Enfermedades de las Cabras/inmunología , Hemoncosis/veterinaria , Haemonchus/inmunología , Intestinos/inmunología , Ganglios Linfáticos/inmunología , Abomaso/parasitología , Animales , Anticuerpos Antihelmínticos/biosíntesis , Anticuerpos Antihelmínticos/sangre , Antígenos Helmínticos/sangre , Linfocitos T CD4-Positivos/inmunología , Ensayo de Inmunoadsorción Enzimática , Enfermedades de las Cabras/patología , Cabras , Hemoncosis/inmunología , Hemoncosis/patología , Inmunización , Intestinos/citología , Ganglios Linfáticos/patología , Activación de Linfocitos , Recuento de Huevos de Parásitos
14.
Blood ; 100(3): 869-78, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12130497

RESUMEN

Using in vitro progenitor assays, serum-free in vitro cultures, and the nonobese diabetic/severe combined immune-deficient (NOD/SCID) ecotropic murine virus knockout xenotransplantation model to detect human SCID repopulating cells (SRCs) with multilineage reconstituting function, we have characterized and compared purified subpopulations harvested from the peripheral blood (PB) of patients receiving granulocyte colony-stimulating factor (G-CSF) alone or in combination with stem cell factor (SCF). Mobilized G-CSF plus SCF PB showed a 2-fold increase in total mononuclear cell content and a 5-fold increase in CD34-expressing cells depleted for lineage-marker expression (CD34(+)Lin(-)) as compared with patients treated with G-CSF alone. Functionally, G-CSF plus SCF-mobilized CD34(+)CD38(-)Lin(-) cells contained a 2-fold enhancement in progenitor frequency as compared with G-CSF-mobilized subsets. Despite enhanced cellularity and progenitor capacity, G-CSF plus SCF mobilization did not increase the frequency of SRCs as determined by limiting dilution analysis by means of unfractionated PB cells. Purification of SRCs from these sources demonstrated that as few as 1000 CD34(+)CD38(-)Lin(-) cells from G-CSF-mobilized PB contained SRC capacity while G-CSF plus SCF-mobilized CD34(+)CD38(-)Lin(-) cells failed to repopulate at doses up to 500 000 cells. In addition, primitive CD34(-)CD38(-)AC133(+)Lin(-) cells derived from G-CSF plus SCF-mobilized PB were capable of differentiation into CD34-expressing cells, while the identical subfractions from G-CSF PB were unable to produce CD34(+) cells in serum-free cultures. Our study defines qualitative and quantitative distinctions among subsets of primitive cells mobilized by means of G-CSF plus SCF versus G-CSF alone, and therefore has implications for the utility of purified repopulating cells from these sources.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética/métodos , Células Madre Hematopoyéticas/efectos de los fármacos , Factor de Células Madre/farmacología , Animales , Antígenos CD34/análisis , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Interacciones Farmacológicas , Hematopoyesis/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Humanos , Inmunofenotipificación , Ratones , Ratones SCID , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...