Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mil Med ; 189(3-4): 80-84, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-37855327

RESUMEN

In response to the Sergeant First Class Heath Robinson Honoring our Promise to Address Comprehensive Toxics (PACT) Act being signed into law, several research groups in Colorado organized the First Annual PACT Act Research Symposium for Veteran Health. The 2-day symposium was interested in research relevant to military exposures with a primary focus on respiratory and mental health. Information on the PACT Act, data sources in the Department of Veteran Affairs and DOD, and research opportunities at the Veteran Affairs were presented. The morning session centered on respiratory health, highlighting research conducted over the last two decades regarding deployment-related respiratory diseases. Despite the high prevalence of mental health disorders among Veterans, information presented during the afternoon sessions on mental health highlighted the dearth of research to date regarding psychological health and military-related exposures. Policymakers, clinicians, and researchers were encouraged to adopt a life-course approach when conceptualizing physical and psychological exposures. On the second day of meetings, a smaller group of participants discussed next steps in military exposure research, as well as priorities for future research. Per the latter, recommendations for future research were made regarding the need for more precise exposure characterization, longitudinal data collection, and efforts to increase understanding regarding disease pathogenesis, as well as the impact of exposures across multiple organs. Such efforts will require interdisciplinary collaboration.


Asunto(s)
Salud de los Veteranos , Veteranos , Estados Unidos , Humanos , Colorado , United States Department of Veterans Affairs , Atención Dirigida al Paciente , Grupo de Atención al Paciente , Lomustina , Prednisona
2.
PLoS One ; 18(2): e0281452, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36757935

RESUMEN

The advent of micro-computed tomography (microCT) has provided significant advancement in our ability to generate clinically relevant assessments of lung health and disease in small animal models. As microCT use to generate outcomes analysis in pulmonary preclinical models has increased there have been substantial improvements in image quality and resolution, and data analysis software. However, there are limited published methods for standardized imaging and automated analysis available for investigators. Manual quantitative analysis of microCT images is complicated by the presence of inflammation and parenchymal disease. To improve the efficiency and limit user-associated bias, we have developed an automated pulmonary air and tissue segmentation (PATS) task list to segment lung air volume and lung tissue volume for quantitative analysis. We demonstrate the effective use of the PATS task list using four distinct methods for imaging, 1) in vivo respiration controlled scanning using a flexiVent, 2) longitudinal breath-gated in vivo scanning in resolving and non-resolving pulmonary disease initiated by lipopolysaccharide-, bleomycin-, and silica-exposure, 3) post-mortem imaging, and 4) ex vivo high-resolution scanning. The accuracy of the PATS task list was compared to manual segmentation. The use of these imaging techniques and automated quantification methodology across multiple models of lung injury and fibrosis demonstrates the broad applicability and adaptability of microCT to various lung diseases and small animal models and presents a significant advance in efficiency and standardization of preclinical microCT imaging and analysis for the field of pulmonary research.


Asunto(s)
Enfermedades Pulmonares , Ratones , Animales , Microtomografía por Rayos X/métodos , Enfermedades Pulmonares/diagnóstico por imagen , Enfermedades Pulmonares/patología , Pulmón/diagnóstico por imagen , Pulmón/patología , Modelos Animales de Enfermedad , Fibrosis
3.
Am J Physiol Lung Cell Mol Physiol ; 320(2): L257-L265, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33355522

RESUMEN

The novel SARS-CoV-2 coronavirus, which is responsible for COVID-19 disease, was first reported in Wuhan, China, in December of 2019. The virus rapidly spread, and the World Health Organization declared a pandemic by March 2020. With millions of confirmed cases worldwide, there is growing concern and considerable debate regarding the potential for coronavirus infection to contribute to an appreciable burden of chronic respiratory symptoms or fibrotic disease among recovered individuals. Because the first case of COVID-19 was documented less than one year ago, data regarding long-term clinical outcomes are not yet available, and predictions for long-term outcome are speculative at best. However, due to the staggering number of cases and the severity of disease in many individuals, there is a critical need to consider the potential long-term implications of COVID-19. This review examines current basic and clinical data regarding fibrogenic mechanisms of viral injury in the context of SARS-CoV-2. Several intersecting mechanisms between coronavirus infection and fibrotic pathways are discussed to highlight factors and processes that may be targetable to improve patient outcome. Reports of post-infection sequelae from previous coronavirus outbreaks are presented toward the goal of improved recognition of potential contributing risk factors for fibrotic disease.


Asunto(s)
COVID-19/complicaciones , Pandemias , Fibrosis Pulmonar/etiología , SARS-CoV-2 , Factores de Edad , Anciano , Anciano de 80 o más Años , COVID-19/epidemiología , COVID-19/virología , Citocinas/fisiología , Interacciones Microbiota-Huesped/fisiología , Humanos , Inflamación/etiología , Inflamación/virología , Fibrosis Pulmonar/virología , Respiración Artificial/efectos adversos , Síndrome de Dificultad Respiratoria/complicaciones , Síndrome de Dificultad Respiratoria/etiología , Factores de Riesgo , SARS-CoV-2/patogenicidad , SARS-CoV-2/fisiología , Transducción de Señal , Sobrevivientes
4.
Am J Physiol Heart Circ Physiol ; 320(2): H604-H612, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33306449

RESUMEN

In human heart failure and in murine hearts with left-ventricular pressure overload (LVPO), increases in fibrosis are associated with increases in myocardial stiffness. Secreted protein acidic and rich in cysteine (SPARC) is shown to be necessary for both cardiac fibrosis and increases in myocardial stiffness in response to LVPO; however, cellular sources of cardiac SPARC are incompletely defined. Irradiation and bone marrow transfer were undertaken to test the hypothesis that SPARC expression by bone marrow-derived cells is an important mediator of fibrosis in LVPO. In recipient SPARC-null mice transplanted with donor wild-type (WT) bone marrow and subjected to LVPO, levels of fibrosis similar to that of WT mice were found despite the lack of SPARC expression by resident cells. In recipient WT mice with donor SPARC-null bone marrow, significantly less fibrosis versus that of WT mice was found despite the expression of SPARC by resident cells. Increases in myocardial stiffness followed a similar pattern to that of collagen deposition. Myocardial macrophages were significantly reduced in SPARC-null mice with LVPO versus that of WT mice. Recipient SPARC-null mice transplanted with donor WT bone marrow exhibited an increase in cardiac macrophages versus that of SPARC-null LVPO and donor WT mice with recipient SPARC-null bone marrow. Expression of vascular cellular adhesion molecule (VCAM), a previously identified binding partner of SPARC, was assessed in all groups and with the exception of WT mice, increases in VCAM immunoreactivity with LVPO were observed. However, no differences in VCAM expression between bone marrow transplant groups were noted. In conclusion, SPARC expression by bone marrow-derived cells was critical for fibrotic deposition of collagen and influenced the expansion of myocardial macrophages in response to LVPO.NEW & NOTEWORTHY Myocardial fibrosis and the resultant increases in LV and myocardial stiffness represent pivotal consequences of chronic pressure overload (PO). In this study, a murine model of cardiac fibrosis induced by PO was used to demonstrate a critical function of SPARC in bone marrow-derived cells that drives cardiac fibrosis and increases in cardiac macrophages.


Asunto(s)
Presión Sanguínea , Cardiomegalia/metabolismo , Miocardio/metabolismo , Osteonectina/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Colágenos Fibrilares/metabolismo , Fibrosis , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Osteonectina/genética , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
5.
Inhal Toxicol ; 32(5): 189-199, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32448007

RESUMEN

Objective: The lungs are uniquely exposed to the external environment. Sand and dust exposures in desert regions are common among deployed soldiers. A significant number of Veterans deployed to the Middle East report development of respiratory disorders and diseases.Materials and methods: Sand collected from Fallujah, Iraq and Kandahar, Afghanistan combat zones was analyzed and compared to a sand sample collected from an historic United States (U.S.) battle region (Fort Johnson, James Island, SC, Civil War battle site). Sand samples were analyzed to determine the physical and elemental characteristics that may have the potential to contribute to development of respiratory disease.Results: Using complementary scanning electron microscopy (SEM) imaging and analysis, and inductively coupled plasma mass spectrometry (ICP-MS), it was determined that Iraq sand contained elevated levels of calcium and first row transition metals versus Afghanistan and U.S. sand. Iraq sand particle texture was smooth and round, and particles were considerably smaller than Afghanistan sand. Afghanistan sand was elevated in rare earth metals versus Iraq or U.S. sands and had sharp edge features and larger particle size than Iraq sand.Conclusions: These data demonstrate significant differences in Iraq and Afghanistan sand particle size and characteristics. Middle East sands contained elevated levels of elements that have been associated with respiratory disease versus control site sand, suggesting the potential of sand/dust storm exposure to promote adverse respiratory symptoms. Data also demonstrate the potential for variation based on geographical region or site of exposure. The data generated provide baseline information that will be valuable in designing future exposure studies.


Asunto(s)
Metales/análisis , Arena/química , Afganistán , Conflictos Armados , Irak , Tamaño de la Partícula , South Carolina , Propiedades de Superficie
6.
Front Psychiatry ; 10: 230, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31068843

RESUMEN

Physiological responses to psychological stressors are protective in acute fight or flight situations; however, there is increasing evidence suggesting the detrimental impact of chronic psychological stress on disease. Chronic stress has been associated with inflammation, poor prognosis, increased morbidity, and poor outcome in many diseases including atherosclerosis, cancer, and pulmonary disease. Given the systemic impact of stress, and the role of the hematopoietic system as a rapid responder to homeostatic insults, we hypothesized that early blood profile changes and biochemical alterations could be detected in a model of chronic stress. To test this hypothesis, a variation of the chronic unpredictable stress (CUS) model was employed. Following 10 days of CUS, C57BL/6 mice exhibited a chronic-stress-associated corticosterone profile. Complete blood count (CBC) revealed mild normochromic, normocytic anemia, and reduced monocyte and lymphocyte count. Serum analysis demonstrated hypoferremia with unchanged total iron binding capacity and serum ferritin levels. These findings are consistent with clinical diagnostic parameters for anemia of chronic disease and indicate that CUS results in significant changes in blood and serum biochemical profile in C57BL/6 mice. These studies identify early changes in blood parameters in response to CUS and identify hematopoietic and biochemical alterations that are often associated with increased morbidity in patients experiencing chronic-stress-associated mental health disease.

7.
Front Psychiatry ; 10: 200, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31024360

RESUMEN

The significant biochemical and physiological effects of psychological stress are beginning to be recognized as exacerbating common diseases, including osteoporosis. This review discusses the current evidence for psychological stress-associated mental health disorders as risk factors for osteoporosis, the mechanisms that may link these conditions, and potential implications for treatment. Traditional, alternative, and adjunctive therapies are discussed. This review is not intended to provide therapeutic recommendations, but, rather, the goal of this review is to delineate potential interactions of psychological stress and osteoporosis and to highlight potential multi-system implications of pharmacological interventions. Review of the current literature identifies several potentially overlapping mechanistic pathways that may be of interest (e.g., glucocorticoid signaling, insulin-like growth factor signaling, serotonin signaling) for further basic and clinical research. Current literature also supports the potential for cross-effects of therapeutics for osteoporosis and mental health disorders. While studies examining a direct link between osteoporosis and chronic psychological stress are limited, the studies reviewed herein suggest that a multi-factorial, personalized approach should be considered for improved patient outcomes in populations experiencing psychological stress, particularly those at high-risk for development of osteoporosis.

8.
Cytotherapy ; 20(11): 1371-1380, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30340982

RESUMEN

BACKGROUND AIMS: Previous studies identified a circulating human osteoblastic population that expressed osteocalcin (OCN), increased following fracture and pubertal growth, and formed mineralized colonies in vitro and bone in vivo. A subpopulation expressed CD34, a hematopoietic/endothelial marker. These findings led to our hypothesis that hematopoietic-derived CD34+OCN+ cells exist in the circulation of mice and are modulated after fracture. METHODS: Flow cytometry was used to identify CD34+OCN+ cells in male B6.SJL-PtprcaPepcb/BoyJ and Vav-Cre/mTmG (VavR) mice. Non-stabilized tibial fractures were created by three-point bend. Fractures were longitudinally imaged by micro-computed tomography, and immunofluorescent staining was used to evaluate CD34+OCN+ cells within fracture callus. AMD3100 (10 mg/kg) was injected subcutaneously for 3 days and the CD34+OCN+ population was evaluated by flow cytometry. RESULTS: Circulating CD34+OCN+ cells were identified in mice and confirmed to be of hematopoietic origin (CD45+; Vav1+) using two mouse models. Both circulating and bone marrow-derived CD34+OCN+ cells peaked three weeks post-non-stabilized tibial fracture, suggesting association with cartilage callus transition to bone and early mineralization. Co-expression of CD34 and OCN in the fracture callus at two weeks post-fracture was observed. By three weeks, there was 2.1-fold increase in number of CD34+OCN+ cells, and these were observed throughout the fracture callus. AMD3100 altered CD34+OCN+ cell levels in peripheral blood and bone marrow. DISCUSSION: Together, these data demonstrate a murine CD34+OCN+ circulating population that may be directly involved in fracture repair. Future studies will molecularly characterize CD34+OCN+ cells, determine mechanisms regulating their contribution, and examine if their number correlates with improved fracture healing outcomes.


Asunto(s)
Antígenos CD34/metabolismo , Curación de Fractura/fisiología , Fracturas Óseas/patología , Osteoblastos/citología , Osteocalcina/metabolismo , Animales , Bencilaminas , Biomarcadores/sangre , Médula Ósea/efectos de los fármacos , Ciclamas , Modelos Animales de Enfermedad , Fracturas Óseas/diagnóstico por imagen , Compuestos Heterocíclicos/farmacología , Ratones Transgénicos , Osteoblastos/efectos de los fármacos , Osteoblastos/patología
9.
Am J Physiol Heart Circ Physiol ; 315(1): H92-H100, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29522370

RESUMEN

Myocardial fibrosis and the resultant increases in left ventricular stiffness represent pivotal consequences of chronic pressure overload (PO) that impact both functional capacity and the rates of morbid and mortal events. However, the time course and cellular mechanisms that underlie PO-induced fibrosis have not been completely defined. Secreted protein acidic and rich in cysteine (SPARC) is a matricellular protein that has been shown to be required for insoluble collagen deposition and increased myocardial stiffness in response to PO in mice. As macrophages are associated with increases in fibrillar collagen, the hypothesis that macrophages represent a source of increased SPARC production in the PO myocardium was tested. The time course of changes in the myocardial macrophage population was compared with changes in procollagen type I mRNA, production of SPARC, fibrillar collagen accumulation, and diastolic stiffness. In PO hearts, mRNA encoding collagen type I was increased at 3 days, whereas increases in levels of total collagen protein did not occur until 1 wk and were followed by increases in insoluble collagen at 2 wk. Increases in muscle stiffness were not detected before increases in insoluble collagen content (>1 wk). Significant increases in myocardial macrophages that coincided with increased SPARC were found but did not coincide with increases in mRNA encoding collagen type I. Furthermore, immunohistochemistry and flow cytometry identified macrophages as a cellular source of SPARC. We conclude that myocardial macrophages play an important role in the time-dependent increases in SPARC that enhance postsynthetic collagen processing, insoluble collagen content, and myocardial stiffness and contribute to the development of fibrosis. NEW & NOTEWORTHY Myocardial fibrosis and the resultant increases in left ventricular and myocardial stiffness represent pivotal consequences of chronic pressure overload. In this study a murine model of cardiac fibrosis induced by pressure overload was used to establish a time course of collagen expression, collagen deposition, and cardiac macrophage expansion.


Asunto(s)
Colágeno/metabolismo , Macrófagos/metabolismo , Miocardio/patología , Osteonectina/metabolismo , Animales , Colágeno/genética , Femenino , Fibrosis , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Osteonectina/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
10.
Ann Vasc Surg ; 46: 193-204, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29107003

RESUMEN

BACKGROUND: Hypertension (HTN) has long been associated with abdominal aortic aneurysm (AAA) development, and these cardiovascular pathologies are biochemically characterized by elevated plasma levels of angiotensin II (AngII) as well as interleukin-6 (IL-6). A biologic relationship between HTN and AAA has not been established, however. Accordingly, the objective of this study was to evaluate whether elevated tension may initiate IL-6 production to accumulate monocyte/macrophages and promote dilation of the abdominal aorta (AA). METHODS: An IL-6 infusion model (4.36 µg/kg/day) was created utilizing an osmotic infusion pump, and after 4 weeks, AA diameter was measured by digital microscopy. The AA was then excised for CD68 immunostaining and flow cytometric analysis with CD11b and F4/80 to identify macrophages. Aortic segments from wild-type mice were suspended on parallel wires in an ex vivo tissue myograph at experimentally derived optimal tension (1.2 g) and in the presence of elevated tension (ET, 1.7 g) for 3 hr, and expression of IL-6 and monocyte chemoattractant protein-1 (MCP-1) was evaluated by quantitative polymerase chain reaction (QPCR). Isolated aortic vascular smooth muscle cells (VSMCs) were subjected to 12% biaxial cyclic stretch or held static (control) for 3 hr (n = 7), and IL-6 and MCP-1 expressions were evaluated by QPCR. RESULTS: Four-week IL-6 infusion resulted in an AA outer diameter that was 72.5 ± 5.6% (P < 0.05) greater than that of control mice, and aortic dilation was accompanied by an accumulation of macrophages in the AA medial layer as defined by an increase in CD68 + staining as well as an increase by flow cytometric quantification of CD11b+/F4/80+ cells. Wild-type AA segments did not respond to ex vivo application of ET but cyclic stretch of isolated VSMCs increased IL-6 (2.03 ± 0.3 fold) and MCP-1 (1.51 ± 0.11 fold) expression compared to static control (P < 0.05). Pretreatment with the selective STAT3 inhibitor WP1066 blunted the response in both cases. Interestingly, AngII did not stimulate expression of IL-6 and MCP-1 above that initiated by tension and again, the response was inhibited by WP1066, supporting an integral role of STAT3 in this pathway. CONCLUSIONS: An IL-6 infusion model can initiate macrophage accumulation as well as aortic dilation, and under conditions of elevated tension, this proinflammatory cytokine can be produced by aortic VSMCs. By activation of STAT3, MCP-1 is expressed to increase media macrophage abundance and create an environment susceptible to dilation. This biomechanical association between HTN and aortic dilation may allow for the identification of novel therapeutic strategies.


Asunto(s)
Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/metabolismo , Presión Arterial , Interleucina-6/metabolismo , Angiotensina II , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Aorta Abdominal/patología , Aorta Abdominal/fisiopatología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/fisiopatología , Antígeno CD11b/metabolismo , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Dilatación Patológica , Modelos Animales de Enfermedad , Femenino , Interleucina-6/genética , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Mecanotransducción Celular , Ratones , Monocitos/metabolismo , Monocitos/patología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fosforilación , Factor de Transcripción STAT3/metabolismo , Estrés Mecánico
11.
PLoS One ; 12(7): e0180724, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28700752

RESUMEN

Micro-injuries associated with chronic inhaled particle exposures are linked with activation of the immune response and are thought to contribute to progression of fibrotic disease. In the pulmonary environment, we have previously demonstrated a heterogeneous population of circulating fibroblast precursors (CFPs), which are defined by expression of the pan-leukocyte marker CD45 and the collagen receptor, discoidin domain receptor-2 (DDR2). This population is derived from the hematopoietic stem cell, expresses collagen, and has a fibroblastic morphology in vitro. Herein, we demonstrate a novel subset of CFPs expressing immune markers CD11b, CD11c, and major histocompatibility complex II (MHC II). The CFP population was skewed toward this immune marker expressing subset in animals with silica-induced pulmonary fibrosis. Data indicate that this CFP subset upregulates co-stimulatory molecules and MHC II expression in response to silica-induced fibrosis in vivo. Functionally, this population was shown to promote T cell skewing away from a Th1 response and toward a pro-inflammatory profile. These studies represent the first direct flow cytometric and functional evaluation of the novel immune marker expressing CFP subset in an exposure-induced model of pulmonary fibrosis. Elucidating the role of this CFP subset may enhance our understanding of the complex immune balance critical to mediating exposures at the pulmonary-host interface and may be a valuable target for the treatment of exposure-induced pulmonary fibrosis.


Asunto(s)
Receptor con Dominio Discoidina 2/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/inmunología , Animales , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Fibroblastos/patología , Antígenos de Histocompatibilidad Clase II/metabolismo , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Fenotipo , Fibrosis Pulmonar/patología , Dióxido de Silicio
12.
Stem Cells Int ; 2015: 159713, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26185498

RESUMEN

Multiple origins, including the bone marrow, have been suggested to contribute to fibroblast populations in the lung. Using bone marrow reconstitution strategies, the present study tested the hypothesis that the bone marrow hematopoietic stem cell (HSC) gives rise to lung tissue fibroblasts in vivo. Data demonstrate that the nonadherent bone marrow fraction is enriched for CD45(+) HSC-derived cells and was able to reconstitute hematopoiesis in lethally irradiated animals. Analysis of peripheral blood and lung tissues from engrafted mice demonstrated the ability of this population to give rise to CD45(+)/Discoidin-Domain Receptor-2(+) (DDR2) circulating fibroblast precursors (CFPs) in blood and fibroblast populations in lung. An HSC origin for lung fibroblasts was confirmed using a novel clonal cell transplantation method in which the bone marrow is reconstituted by a clonal population derived from a single HSC. Together, these findings provide evidence for an HSC contribution to lung fibroblasts and demonstrate a circulating intermediate through the CD45(+)/DDR2(+) HSC-derived CFP.

13.
Neoplasia ; 17(5): 434-48, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26025666

RESUMEN

Targeting the tumor microenvironment is critical toward improving the effectiveness of cancer therapeutics. Cancer-associated fibroblasts (CAFs) are one of the most abundant cell types of the tumor microenvironment, playing an important role in tumor progression. Multiple origins for CAFs have been proposed including resident fibroblasts, adipocytes, and bone marrow. Our laboratory previously identified a novel hematopoietic stem cell (HSC) origin for CAFs; however, the functional roles of HSC-derived CAFs (HSC-CAFs) in tumor progression have not yet been examined. To test the hypothesis that HSC-CAFs promote tumor progression through contribution to extracellular matrix (ECM) and paracrine production of pro-angiogenic factors, we developed a method to isolate HSC-CAFs. HSC-CAFs were profiled on the basis of their expression of hematopoietic and fibroblastic markers in two murine tumor models. Profiling revealed production of factors associated with ECM deposition and remodeling. Functional in vivo studies showed that co-injection of HSC-CAFs with tumor cells resulted in increased tumor growth rate and significantly larger tumors than tumor cells alone. Immunohistochemical studies revealed increased blood vessel density with co-injection, demonstrating a role for HSC-CAFs in tumor vascularization. Mechanistic in vitro studies indicated that HSC-CAFs play a role in producing vascular endothelial growth factor A and transforming growth factor-ß1 in endothelial tube formation and patterning. In vitro and in vivo findings suggest that HSC-CAFs are a critical component of the tumor microenvironment and suggest that targeting the novel HSC-CAF may be a promising therapeutic strategy.


Asunto(s)
Fibroblastos/patología , Neoplasias Experimentales/patología , Neovascularización Patológica/patología , Microambiente Tumoral/fisiología , Animales , Western Blotting , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células Madre Hematopoyéticas/patología , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Venas Umbilicales
14.
World J Stem Cells ; 7(2): 253-65, 2015 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-25815113

RESUMEN

The tumor microenvironment (TME) is complex and constantly evolving. This is due, in part, to the crosstalk between tumor cells and the multiple cell types that comprise the TME, which results in a heterogeneous population of tumor cells and TME cells. This review will focus on two stromal cell types, the cancer-associated adipocyte (CAA) and the cancer-associated fibroblast (CAF). In the clinic, the presence of CAAs and CAFs in the TME translates to poor prognosis in multiple tumor types. CAAs and CAFs have an activated phenotype and produce growth factors, inflammatory factors, cytokines, chemokines, extracellular matrix components, and proteases in an accelerated and aberrant fashion. Through this activated state, CAAs and CAFs remodel the TME, thereby driving all aspects of tumor progression, including tumor growth and survival, chemoresistance, tumor vascularization, tumor invasion, and tumor cell metastasis. Similarities in the tumor-promoting functions of CAAs and CAFs suggest that a multipronged therapeutic approach may be necessary to achieve maximal impact on disease. While CAAs and CAFs are thought to arise from tissues adjacent to the tumor, multiple alternative origins for CAAs and CAFs have recently been identified. Recent studies from our lab and others suggest that the hematopoietic stem cell, through the myeloid lineage, may serve as a progenitor for CAAs and CAFs. We hypothesize that the multiple origins of CAAs and CAFs may contribute to the heterogeneity seen in the TME. Thus, a better understanding of the origin of CAAs and CAFs, how this origin impacts their functions in the TME, and the temporal participation of uniquely originating TME cells may lead to novel or improved anti-tumor therapeutics.

15.
J Bone Marrow Res ; 3(3)2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26798846

RESUMEN

The understanding of bone marrow stem cell plasticity and contribution of bone marrow stem cells to pathophysiology is evolving with the advent of innovative technologies. Recent data has led to new mechanistic insights in the field of mesenchymal stem cell (MSC) research, and an increased appreciation for the plasticity of the hematopoietic stem cell (HSC). In this review, we discuss current research examining the origin of pulmonary cell types from endogenous lung stem and progenitor cells as well as bone marrow-derived stem cells (MSCs and HSCs) and their contributions to lung homeostasis and pathology. We specifically highlight recent findings from our laboratory that demonstrate an HSC origin for pulmonary fibroblasts based on transplantation of a clonal population of cells derived from a single HSC. These findings demonstrate the importance of developing an understanding of the sources of effector cells in disease state. Finally, a perspective is given on the potential clinical implications of these studies and others addressing stem cell contributions to lung tissue homeostasis and pathology.

16.
Int J Clin Exp Pathol ; 5(9): 863-73, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23119103

RESUMEN

Researchers have begun to appreciate the significant role that the microenvironment plays in tumorigenesis and are now shedding light on the role of the stroma in induction and progression of solid tumors. While the stroma of solid tumors is comprised of many cell types, including vascular and immune cells, one of the most prominent cell types in the tumor stroma is the fibroblast, called the carcinoma-associated fibroblast (CAF) or tumor-associated fibroblast (TAF). The interaction between CAFs and tumor cells is quite complex. CAFs have been implicated in tumor angiogenesis, immunosuppression, tumor cell proliferation and aggressiveness, genetic instability, and metastasis. However, their specific roles in each of these processes have only been partially elucidated. Determining the role of CAFs has been complicated by the fact that researchers have demonstrated heterogeneity in the stromal fibroblast population. This heterogeneity has brought about the concept of multiple origins for CAFs. While many origins of CAFs have been suggested, in our own laboratory we have identified a novel hematopoietic stem cell (HSC) origin of CAFs. Given the profound role of CAFs in tumor progression and prognosis, the CAF represents an exciting potential therapeutic target. The heterogeneity of the CAF population makes research directed at investigating the roles and origins of CAFs critical to development of such anti-tumor therapies.


Asunto(s)
Carcinoma/patología , Linaje de la Célula , Fibroblastos/patología , Células Madre Hematopoyéticas/patología , Células Madre Neoplásicas/patología , Animales , Biomarcadores de Tumor/metabolismo , Carcinoma/metabolismo , Comunicación Celular , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Fibroblastos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Invasividad Neoplásica , Células Madre Neoplásicas/metabolismo , Microambiente Tumoral
17.
Mol Cell Biol ; 30(22): 5364-80, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20837710

RESUMEN

Murine embryonic stem (ES) cells are defined by continuous self-renewal and pluripotency. A diverse repertoire of protein isoforms arising from alternative splicing is expressed in ES cells without defined biological roles. Sall4, a transcription factor essential for pluripotency, exists as two isoforms (Sall4a and Sall4b). Both isoforms can form homodimers and a heterodimer with each other, and each can interact with Nanog. By genomewide location analysis, we determined that Sall4a and Sall4b have overlapping, but not identical binding sites within the ES cell genome. In addition, Sall4b, but not Sall4a, binds preferentially to highly expressed loci in ES cells. Sall4a and Sall4b binding sites are distinguished by both epigenetic marks at target loci and their clustering with binding sites of other pluripotency factors. When ES cells expressing a single isoform of Sall4 are generated, Sall4b alone could maintain the pluripotent state, although it could not completely suppress all differentiation markers. Sall4a and Sall4b collaborate in maintenance of the pluripotent state but play distinct roles. Our work is novel in establishing such isoform-specific differences in ES cells.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes/fisiología , Isoformas de Proteínas/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Dimerización , Epigénesis Genética , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Análisis por Micromatrices , Datos de Secuencia Molecular , Proteína Homeótica Nanog , Células Madre Pluripotentes/citología , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estructura Cuaternaria de Proteína , Factores de Transcripción/química , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...