Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Curr Biol ; 33(18): 3821-3834.e5, 2023 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-37572663

RESUMO

During central nervous system (CNS) development, a precisely patterned vasculature emerges to support CNS function. How neurons control angiogenesis is not well understood. Here, we show that the neuromodulator dopamine restricts vascular development in the retina via temporally limited production by an unexpected neuron subset. Our genetic and pharmacological experiments demonstrate that elevating dopamine levels inhibits tip-cell sprouting and vessel growth, whereas reducing dopamine production by all retina neurons increases growth. Dopamine production by canonical dopaminergic amacrine interneurons is dispensable for these events. Instead, we found that temporally restricted dopamine production by retinal ganglion cells (RGCs) modulates vascular development. RGCs produce dopamine precisely during angiogenic periods. Genetically limiting dopamine production by ganglion cells, but not amacrines, decreases angiogenesis. Conversely, elevating ganglion-cell-derived dopamine production inhibits early vessel growth. These vasculature outcomes occur downstream of vascular endothelial growth factor receptor (VEGFR) activation and Notch-Jagged1 signaling. Jagged1 is increased and subsequently inhibits Notch signaling when ganglion cell dopamine production is reduced. Our findings demonstrate that dopaminergic neural activity from a small neuron subset functions upstream of VEGFR to serve as developmental timing cue that regulates vessel growth.


Assuntos
Dopamina , Fator A de Crescimento do Endotélio Vascular , Dopamina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Retina , Células Ganglionares da Retina/metabolismo , Transdução de Sinais
2.
Nat Commun ; 14(1): 1929, 2023 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-37024491

RESUMO

Activating non-inherited mutations in the guanine nucleotide-binding protein G(q) subunit alpha (GNAQ) gene family have been identified in childhood vascular tumors. Patients experience extensive disfigurement, chronic pain and severe complications including a potentially lethal coagulopathy termed Kasabach-Merritt phenomenon. Animal models for this class of vascular tumors do not exist. This has severely hindered the discovery of the molecular consequences of GNAQ mutations in the vasculature and, in turn, the preclinical development of effective targeted therapies. Here we report a mouse model expressing hyperactive mutant GNAQ in endothelial cells. Mutant mice develop vascular and coagulopathy phenotypes similar to those seen in patients. Mechanistically, by transcriptomic analysis we demonstrate increased mitogen activated protein kinase signaling in the mutant endothelial cells. Targeting of this pathway with Trametinib suppresses the tumor growth by reducing vascular cell proliferation and permeability. Trametinib also prevents the development of coagulopathy and improves mouse survival.


Assuntos
Melanoma , Neoplasias Uveais , Neoplasias Vasculares , Animais , Camundongos , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Células Endoteliais/metabolismo , Apoptose , Melanoma/genética , Neoplasias Uveais/genética , Mutação , Modelos Animais de Doenças , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Linhagem Celular Tumoral
3.
Toxicol Pathol ; 50(7): 836-857, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36165586

RESUMO

The 2022 annual National Toxicology Program Satellite Symposium, entitled "Pathology Potpourri," was held in Austin, Texas at the Society of Toxicologic Pathology's 40th annual meeting during a half-day session on Sunday, June 19. The goal of this symposium was to present and discuss challenging diagnostic pathology and/or nomenclature issues. This article presents summaries of the speakers' talks along with select images that were used by the audience for voting and discussion. Various lesions and topics covered during the symposium included induced and spontaneous neoplastic and nonneoplastic lesions in the mouse lung, spontaneous lesions in the reproductive tract of a female cynomolgus macaque, induced vascular lesions in a mouse asthma model and interesting case studies in a rhesus macaque, dog and genetically engineered mouse model.


Assuntos
Toxicologia , Camundongos , Feminino , Animais , Cães , Macaca mulatta , Macaca fascicularis
4.
Int J Radiat Oncol Biol Phys ; 113(3): 661-674, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35361520

RESUMO

PURPOSE: Cancer is a severe delayed effect of acute radiation exposure. Total-body irradiation has been associated with an increased risk of solid cancer and leukemia in Japanese atomic bomb survivors, and secondary malignancies, such as sarcoma, are a serious consequence of cancer radiation therapy. The radiation late effects cohort (RLEC) of rhesus macaques (Macaca mulatta) is a unique resource of more than 200 animals for studying the long-term consequences of total-body irradiation in an animal model that closely resembles humans at the genetic and physiologic levels. METHODS AND MATERIALS: Using clinical records, clinical imaging, histopathology, and immunohistochemistry, this retrospective study characterized the incidence of neoplasia in the RLEC. RESULTS: Since 2007, 61 neoplasms in 44 of 239 irradiated animals were documented (18.4% of the irradiated population). Only 1 neoplasm was diagnosed among the 51 nonirradiated controls of the RLEC (2.0%). The most common malignancies in the RLEC were sarcomas (38.3% of diagnoses), which are rare neoplasms in nonirradiated macaques. The most common sarcomas included malignant nerve sheath tumors and malignant glomus tumors. Carcinomas were less common (19.7% of diagnoses), and consisted primarily of renal cell and hepatocellular carcinomas. Neoplasia occurred in most major body systems, with the skin and subcutis being the most common site (40%). RNA analysis showed similarities in transcriptional profiles between RLEC and human malignant nerve sheath tumors. CONCLUSIONS: This study indicates that total-body irradiation is associated with an increased incidence of neoplasia years following irradiation, at more than double the incidence described in aging, nonirradiated animals, and promotes tumor histotypes that are rarely observed in nonirradiated, aging rhesus macaques.


Assuntos
Neoplasias de Bainha Neural , Lesões por Radiação , Sarcoma , Animais , Humanos , Incidência , Macaca mulatta , Estudos Retrospectivos , Sarcoma/epidemiologia , Sarcoma/etiologia , Sarcoma/veterinária
5.
J Neurosci ; 41(50): 10247-10260, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34759029

RESUMO

Axon regeneration after spinal cord injury (SCI) is limited by both a decreased intrinsic ability of neurons to grow axons and the growth-hindering effects of extrinsic inhibitory molecules expressed around the lesion. Deletion of phosphatase and tensin homolog (Pten) augments mechanistic target of rapamycin (mTOR) signaling and enhances the intrinsic regenerative response of injured corticospinal neurons after SCI. Because of the variety of growth-restrictive extrinsic molecules, it remains unclear how inhibition of conserved inhibitory signaling elements would affect axon regeneration and rewiring after SCI. Moreover, it remains unknown how a combinatorial approach to modulate both extrinsic and intrinsic mechanisms can enhance regeneration and rewiring after SCI. In the present study, we deleted RhoA and RhoC, which encode small GTPases that mediate growth inhibition signals of a variety of extrinsic molecules, to remove global extrinsic pathways. RhoA/RhoC double deletion in mice suppressed retraction or dieback of corticospinal axons after SCI. In contrast, Pten deletion increased regrowth of corticospinal axons into the lesion core. Although deletion of both RhoA and Pten did not promote axon regrowth across the lesion or motor recovery, it additively promoted rewiring of corticospinal circuits connecting the cerebral cortex, spinal cord, and hindlimb muscles. Our genetic findings, therefore, reveal that a combinatorial approach to modulate both intrinsic and extrinsic factors can additively promote neural circuit rewiring after SCI.SIGNIFICANCE STATEMENT SCI often causes severe motor deficits because of damage to the corticospinal tract (CST), the major neural pathway for voluntary movements. Regeneration of CST axons is required to reconstruct motor circuits and restore functions; however, a lower intrinsic ability to grow axons and extrinsic inhibitory molecules severely limit axon regeneration in the CNS. Here, we investigated whether suppression of extrinsic inhibitory cues by genetic deletion of Rho as well as enhancement of the intrinsic pathway by deletion of Pten could enable axon regrowth and rewiring of the CST after SCI. We show that simultaneous elimination of extrinsic and intrinsic signaling pathways can additively promote axon sprouting and rewiring of the corticospinal circuits. Our data demonstrate a potential molecular approach to reconstruct motor pathways after SCI.


Assuntos
Regeneração Nervosa/fisiologia , PTEN Fosfo-Hidrolase/metabolismo , Tratos Piramidais/fisiopatologia , Traumatismos da Medula Espinal/fisiopatologia , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34031241

RESUMO

Myopia has become a major public health concern, particularly across much of Asia. It has been shown in multiple studies that outdoor activity has a protective effect on myopia. Recent reports have shown that short-wavelength visible violet light is the component of sunlight that appears to play an important role in preventing myopia progression in mice, chicks, and humans. The mechanism underlying this effect has not been understood. Here, we show that violet light prevents lens defocus-induced myopia in mice. This violet light effect was dependent on both time of day and retinal expression of the violet light sensitive atypical opsin, neuropsin (OPN5). These findings identify Opn5-expressing retinal ganglion cells as crucial for emmetropization in mice and suggest a strategy for myopia prevention in humans.


Assuntos
Cristalino/metabolismo , Luz , Proteínas de Membrana/metabolismo , Miopia/prevenção & controle , Opsinas/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Miopia/metabolismo , Refração Ocular , Tomografia de Coerência Óptica , Corpo Vítreo
7.
Am J Respir Cell Mol Biol ; 64(1): 59-68, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33058732

RESUMO

Recently, we characterized blue light-mediated relaxation (photorelaxation) of airway smooth muscle (ASM) and implicated the involvement of opsin 3 (OPN3), an atypical opsin. In the present study, we characterized the cellular signaling mechanisms of photorelaxation. We confirmed the functional role of OPN3 in blue light photorelaxation using trachea from OPN3 null mice (maximal relaxation 52 ± 13% compared with wild-type mice 90 ± 4.3%, P < 0.05). We then demonstrated colocalization of OPN3 and Gαs using co-IP and proximity ligation assays in primary human ASM cells, which was further supported by an increase in cAMP in mouse trachea treated with blue light compared with dark controls (23 ± 3.6 vs. 14 ± 2.6 pmol cAMP/ring, P < 0.05). Downstream PKA (protein kinase A) involvement was shown by inhibiting photorelaxation using Rp-cAMPS (P < 0.0001). Moreover, we observed converging mechanisms of desensitization by chronic ß2-agonist exposure in mouse trachea and correlated this finding with colocalization of OPN3 and GRK2 (G protein receptor kinase) in primary human ASM cells. Finally, an overexpression model of OPN1LW (a red light photoreceptor in the same opsin family) in human ASM cells showed an increase in intracellular cAMP levels following red light exposure compared with nontransfected cells (48 ± 13 vs. 13 ± 2.1 pmol cAMP/mg protein, P < 0.01), suggesting a conserved photorelaxation mechanism for wavelengths of light that are more tissue penetrant. Together, these results demonstrate that blue light photorelaxation in ASM is mediated by the OPN3 receptor interacting with Gαs, which increases cAMP levels, activating PKA and modulated by GRK2.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Relaxamento Muscular/fisiologia , Músculo Liso/metabolismo , Miócitos de Músculo Liso/metabolismo , Opsinas de Bastonetes/metabolismo , Traqueia/metabolismo , Animais , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Opsinas/metabolismo , Transdução de Sinais/fisiologia
8.
Nature ; 585(7825): 420-425, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32879486

RESUMO

The opsin family of G-protein-coupled receptors are used as light detectors in animals. Opsin 5 (also known as neuropsin or OPN5) is a highly conserved opsin that is sensitive to visible violet light1,2. In mice, OPN5 is a known photoreceptor in the retina3 and skin4 but is also expressed in the hypothalamic preoptic area (POA)5. Here we describe a light-sensing pathway in which POA neurons that express Opn5 regulate thermogenesis in brown adipose tissue (BAT). We show that Opn5 is expressed in glutamatergic warm-sensing POA neurons that receive synaptic input from several thermoregulatory nuclei. We further show that Opn5 POA neurons project to BAT and decrease its activity under chemogenetic stimulation. Opn5-null mice show overactive BAT, increased body temperature, and exaggerated thermogenesis when cold-challenged. Moreover, violet photostimulation during cold exposure acutely suppresses BAT temperature in wild-type mice but not in Opn5-null mice. Direct measurements of intracellular cAMP ex vivo show that Opn5 POA neurons increase cAMP when stimulated with violet light. This analysis thus identifies a violet light-sensitive deep brain photoreceptor that normally suppresses BAT thermogenesis.


Assuntos
Cor , Luz , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Neurônios/efeitos da radiação , Opsinas/metabolismo , Área Pré-Óptica/citologia , Termogênese/efeitos da radiação , Tecido Adiposo Marrom/inervação , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Marrom/efeitos da radiação , Animais , Temperatura Corporal , Temperatura Baixa , AMP Cíclico/metabolismo , Feminino , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Opsinas/deficiência , Opsinas/genética , Termogênese/genética
9.
Neurourol Urodyn ; 39(6): 1667-1678, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32531084

RESUMO

AIMS: As PDGFRα (+) cells appear not to suppress the excitability of detrusor smooth muscle by generating SK3-dependent hyperpolarising as proposed in the gastrointestinal tract, we further explored the functional roles of PDGFRα (+) cells in regulating the spontaneous activity of urogenital tissues. METHODS: Using PDGFRα-eGFP mice, intracellular Ca2+ signaling in PDGFRα (+) cells of the bladder lamina propria, renal pelvis, and seminal vesicle were visualized using Cal-590 fluorescence. The distribution and SK3 expression of PDGFRα (+) cells were also examined by immunohistochemistry. RESULTS: In the bladder lamina propria, SK3 (-) PDGFRα (+) cells exhibited spontaneous Ca2+ transients and responded to stimulation of P2Y1 purinoceptors with MRS2365 (100 nM) or adenosine diphosphate (ADP) (100 µM) by developing Ca2+ transients. In the proximal renal pelvis, PDGFRα (+) cells were distributed in the mucosal, muscular and serosal layers but did not express SK3 immunoreactivity. PDGFRα (+) cells in the musculature resembling atypical smooth muscle cells generated spontaneous Ca2+ transients that were partially suppressed upon P2Y1-stimulation, while vigorously responding to human angiotensin II (100 nM). In the seminal vesicle, PDGFRα (+) cells in the musculature but not mucosa expressed SK3 immunoreactivity. In the mucosa, the P2Y1 stimulation evoked Ca2+ transients in both PDGFRα (+) cells and PDGFRα (-) cells. CONCLUSION: PDGFRα (+) cells in spontaneously active urogenital tissues display heterogeneity in terms of their SK3 expression and P2Y1-induced Ca2+ responses. Muscular PDGFRα (+) cells in the renal pelvis and mucosal PDGFRα (+) cells in the seminal vesicle may generate depolarizing signals to drive smooth muscle cells.


Assuntos
Músculo Liso/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Bexiga Urinária/metabolismo , Difosfato de Adenosina/análogos & derivados , Animais , Masculino , Camundongos , Camundongos Transgênicos , Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Bexiga Urinária/diagnóstico por imagem
10.
Development ; 145(12)2018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29777010

RESUMO

Normal development requires tight regulation of cell proliferation and cell death. Here, we have investigated these control mechanisms in the hyaloid vessels, a temporary vascular network in the mammalian eye that requires a Wnt/ß-catenin response for scheduled regression. We investigated whether the hyaloid Wnt response was linked to the oncogene Myc, and the cyclin-dependent kinase inhibitor CDKN1A (P21), both established regulators of cell cycle progression and cell death. Our analysis showed that the Wnt pathway co-receptors LRP5 and LRP6 have overlapping activities that mediate the Wnt/ß-catenin signaling in hyaloid vascular endothelial cells (VECs). We also showed that both Myc and Cdkn1a are downstream of the Wnt response and are required for hyaloid regression but for different reasons. Conditional deletion of Myc in VECs suppressed both proliferation and cell death. By contrast, conditional deletion of Cdkn1a resulted in VEC overproliferation that countered the effects of cell death on regression. When combined with analysis of MYC and CDKN1A protein levels, this analysis suggests that a Wnt/ß-catenin and MYC-CDKN1A pathway regulates scheduled hyaloid vessel regression.


Assuntos
Apoptose/fisiologia , Proliferação de Células/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Endotélio Vascular/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , beta Catenina/metabolismo , Animais , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21/genética , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Olho/irrigação sanguínea , Células HEK293 , Humanos , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-myc/genética , Via de Sinalização Wnt/fisiologia
11.
Ophthalmic Surg Lasers Imaging Retina ; 48(12): 962-968, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29253298

RESUMO

BACKGROUND AND OBJECTIVE: To report clinical feasibility of non-mydriatic ultra-widefield (NMUWF) imaging and determine the prevalence of peripheral retinal pathology in comparison to standard single-field imaging in a primary care setting. PATIENTS AND METHODS: Six hundred and thirty-two subjects (1,260 eyes) who underwent NMUWF imaging during annual health screening from October 2015 through March 2016 were retrospectively identified. An automated algorithm processed the raw images into: (1) NMUWF image with mask/grid outline that delineates the center 45° field simulating standard single-field photograph and (2) single-field image comprising 45° posterior pole extracted from the corresponding NMUWF image. RESULTS: Mean age of patients was 59.6 years ± 7.5 years. Of the 1,260 eyes, 1,238 eyes (98.3%) were considered optimum for grading. NMUWF images detected peripheral retinal pathology in 228 eyes (18.4%) that were not visible on corresponding single-field images. CONCLUSIONS: NMUWF imaging is feasible in a primary care setting, allows improved visualization of peripheral retinal pathology, and may therefore be useful for telemedicine screening. [Ophthalmic Surg Lasers Imaging Retina. 2017;48:962-968.].


Assuntos
Angiofluoresceinografia/métodos , Oftalmoscopia/métodos , Fotografação/métodos , Retina/patologia , Doenças Retinianas/diagnóstico , Algoritmos , Estudos de Viabilidade , Feminino , Fundo de Olho , Humanos , Masculino , Pessoa de Meia-Idade , Midriáticos , Reprodutibilidade dos Testes , Estudos Retrospectivos
12.
J Trauma Nurs ; 24(5): 306-311, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28885519

RESUMO

Trauma leads to 5.7 million annual deaths globally, accounting for 25%-33% of global unintentional deaths and 90% of the global trauma burden in low- and middle-income countries. The Lancet Commission on Global Surgery and the World Health Organization assert that emergent and essential surgical capacity building and trauma system improvement are essential to address the global burden of trauma. In response, the Rutgers Global Surgery program, the School of Nursing and Medicine, and the Robert Wood Johnson University Hospital faculty collaborated in the first Interprofessional Models in Global Injury Care and Education Symposium in June 2016. This 2-week symposium combined lectures, high-fidelity simulation, small group workshops, site visits to Level I trauma centers, and a 1-day training course from the Panamerican Trauma Society. The aim was to introduce global trauma nurses to trauma leadership and trauma system development. After completing the symposium, 10 nurses from China, Colombia, Kenya, Puerto Rico, and Uruguay were surveyed. Overall, 88.8% of participants reported high levels of satisfaction with the program and 100% stated being very satisfied with trauma lectures. Symposia, such as that developed and offered by Rutgers University, prepare nurses to address trauma within system-based care and facilitate trauma nursing leadership in their respective countries.


Assuntos
Educação em Enfermagem/métodos , Liderança , Papel do Profissional de Enfermagem , Centros de Traumatologia/organização & administração , Ferimentos e Lesões/enfermagem , China , Colômbia , Feminino , Saúde Global , Humanos , Quênia , Masculino , Enfermagem , Porto Rico , Uruguai
13.
PLoS One ; 12(8): e0181549, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28846685

RESUMO

Resident tissue myeloid cells play a role in many aspects of physiology including development of the vascular systems. In the blood vasculature, myeloid cells use VEGFC to promote angiogenesis and can use Wnt ligands to control vascular branching and to promote vascular regression. Here we show that myeloid cells also regulate development of the dermal lymphatic vasculature using Wnt ligands. Using myeloid-specific deletion of the WNT transporter Wntless we show that myeloid Wnt ligands are active at two distinct stages of development of the dermal lymphatics. As lymphatic progenitors are emigrating from the cardinal vein and intersomitic vessels, myeloid Wnt ligands regulate both their numbers and migration distance. Later in lymphatic development, myeloid Wnt ligands regulate proliferation of lymphatic endothelial cells (LEC) and thus control lymphatic vessel caliber. Myeloid-specific deletion of WNT co-receptor Lrp5 or Wnt5a gain-of-function also produce elevated caliber in dermal lymphatic capillaries. These data thus suggest that myeloid cells produce Wnt ligands to regulate lymphatic development and use Wnt pathway co-receptors to regulate the balance of Wnt ligand activity during the macrophage-LEC interaction.


Assuntos
Derme/metabolismo , Células Endoteliais/metabolismo , Linfangiogênese/fisiologia , Vasos Linfáticos/metabolismo , Células Mieloides/metabolismo , Proteínas Wnt/metabolismo , Animais , Proliferação de Células/fisiologia , Camundongos , Via de Sinalização Wnt
14.
J Smooth Muscle Res ; 53(0): 57-72, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28652517

RESUMO

The prostate is a gland whose secretions contribute to the seminal fluids ejaculated upon activation of autonomic sympathetic nerves. In elder males, the prostate undergoes an increase in stroma mass and myogenic tone, leading to benign prostatic hyperplasia that occludes the proximal urethra and the presentation of various lower urinary tract symptoms that decrease their quality of life. This review summarises the role of prostatic interstitial cells (PICs) in the generation of the spontaneous tone in the prostate. It presents current knowledge of the role of Ca2+ plays in PIC pacemaking, as well as the mechanisms by which this spontaneous activity triggers slow wave generation and stromal contraction. PICs display a small T-type Ca2+ current (ICaT) and a large L-type Ca2+ current (ICaL). In contrast to other interstitial cells in the urinary and gastrointestinal tracts, spontaneous Ca2+ signalling in PICs is uniquely dependent on Ca2+ influx through ICaL channels. A model of prostatic pacemaking is presented describing how ICaL can be triggered by an initial membrane depolarization evoked upon the selective opening of Ca2+-activated Cl- channels by Ca2+ flowing only through ICaT channels. The resulting current flow through ICaL results in release of Ca2+ from internal stores and the summation of Cl--selective spontaneous transient depolarizations (STDs) to form pacemaker potentials that propagate passively into the prostatic stroma to evoke regenerative action potentials and excitation-contraction coupling.


Assuntos
Próstata/citologia , Próstata/fisiologia , Potenciais de Ação/fisiologia , Animais , Relógios Biológicos/fisiologia , Cálcio/metabolismo , Cálcio/fisiologia , Canais de Cálcio/fisiologia , Sinalização do Cálcio/fisiologia , Canais de Cloreto/fisiologia , Fenômenos Eletrofisiológicos , Gerbillinae , Cobaias , Humanos , Técnicas In Vitro , Canais Iônicos/fisiologia , Masculino , Camundongos , Hiperplasia Prostática/etiologia , Ratos , Sistema Nervoso Simpático/fisiologia , Transmissão Sináptica/fisiologia
15.
J Am Heart Assoc ; 6(1)2017 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-28062479

RESUMO

BACKGROUND: Macrophages and Wnt proteins (Wnts) are independently involved in cardiac development, response to cardiac injury, and repair. However, the role of macrophage-derived Wnts in the healing and repair of myocardial infarction (MI) is unknown. We sought to determine the role of macrophage Wnts in infarct repair. METHODS AND RESULTS: We show that the Wnt pathway is activated after MI in mice. Furthermore, we demonstrate that isolated infarct macrophages express distinct Wnt pathway components and are a source of noncanonical Wnts after MI. To determine the effect of macrophage Wnts on cardiac repair, we evaluated mice lacking the essential Wnt transporter Wntless (Wls) in myeloid cells. Significantly, Wntless-deficient macrophages presented a unique subset of M2-like macrophages with anti-inflammatory, reparative, and angiogenic properties. Serial echocardiography studies revealed that mice lacking macrophage Wnt secretion showed improved function and less remodeling 30 days after MI. Finally, mice lacking macrophage-Wntless had increased vascularization near the infarct site compared with controls. CONCLUSIONS: Macrophage-derived Wnts are implicated in adverse cardiac remodeling and dysfunction after MI. Together, macrophage Wnts could be a new therapeutic target to improve infarct healing and repair.


Assuntos
Coração/diagnóstico por imagem , Peptídeos e Proteínas de Sinalização Intracelular/genética , Macrófagos/metabolismo , Infarto do Miocárdio/diagnóstico por imagem , Neovascularização Fisiológica/genética , Receptores Acoplados a Proteínas G/genética , Remodelação Ventricular/genética , Proteínas Wnt/metabolismo , Animais , Modelos Animais de Doenças , Ecocardiografia , Feminino , Macrófagos/imunologia , Camundongos , Infarto do Miocárdio/imunologia , Neovascularização Fisiológica/imunologia , Remodelação Ventricular/imunologia , Via de Sinalização Wnt
17.
Development ; 143(2): 356-66, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26681494

RESUMO

The developing lens is a powerful system for investigating the molecular basis of inductive tissue interactions and for studying cataract, the leading cause of blindness. The formation of tightly controlled cell-cell adhesions and cell-matrix junctions between lens epithelial (LE) cells, between lens fiber (LF) cells, and between these two cell populations enables the vertebrate lens to adopt a highly ordered structure and acquire optical transparency. Adhesion molecules are thought to maintain this ordered structure, but little is known about their identity or interactions. Cysteine-rich motor neuron 1 (Crim1), a type I transmembrane protein, is strongly expressed in the developing lens and its mutation causes ocular disease in both mice and humans. How Crim1 regulates lens morphogenesis is not understood. We identified a novel ENU-induced hypomorphic allele of Crim1, Crim1(glcr11), which in the homozygous state causes cataract and microphthalmia. Using this and two other mutant alleles, Crim1(null) and Crim1(cko), we show that the lens defects in Crim1 mouse mutants originate from defective LE cell polarity, proliferation and cell adhesion. Crim1 adhesive function is likely to be required for interactions both between LE cells and between LE and LF cells. We show that Crim1 acts in LE cells, where it colocalizes with and regulates the levels of active ß1 integrin and of phosphorylated FAK and ERK. The RGD and transmembrane motifs of Crim1 are required for regulating FAK phosphorylation. These results identify an important function for Crim1 in the regulation of integrin- and FAK-mediated LE cell adhesion during lens development.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Cristalino/citologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas/genética , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Cristalino/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Organogênese/genética , Organogênese/fisiologia , Fosforilação , Transdução de Sinais/fisiologia
18.
J Exp Med ; 212(9): 1433-48, 2015 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-26261265

RESUMO

Although the link between inflammation and cancer initiation is well established, its role in metastatic diseases, the primary cause of cancer deaths, has been poorly explored. Our previous studies identified a population of metastasis-associated macrophages (MAMs) recruited to the lung that promote tumor cell seeding and growth. Here we show that FMS-like tyrosine kinase 1 (Flt1, also known as VEGFR1) labels a subset of macrophages in human breast cancers that are significantly enriched in metastatic sites. In mouse models of breast cancer pulmonary metastasis, MAMs uniquely express FLT1. Using several genetic models, we show that macrophage FLT1 signaling is critical for metastasis. FLT1 inhibition does not affect MAM recruitment to metastatic lesions but regulates a set of inflammatory response genes, including colony-stimulating factor 1 (CSF1), a central regulator of macrophage biology. Using a gain-of-function approach, we show that CSF1-mediated autocrine signaling in MAMs is downstream of FLT1 and can restore the tumor-promoting activity of FLT1-inhibited MAMs. Thus, CSF1 is epistatic to FLT1, establishing a link between FLT1 and inflammatory responses within breast tumor metastases. Importantly, FLT1 inhibition reduces tumor metastatic efficiency even after initial seeding, suggesting that these pathways represent therapeutic targets in metastatic disease.


Assuntos
Neoplasias da Mama/metabolismo , Macrófagos/metabolismo , Neoplasias Mamárias Animais/metabolismo , Proteínas de Neoplasias/metabolismo , Transdução de Sinais , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Comunicação Autócrina/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Humanos , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/patologia , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Transgênicos , Metástase Neoplásica , Proteínas de Neoplasias/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
19.
Dis Model Mech ; 8(8): 969-76, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26035379

RESUMO

The belly spot and tail (Bst(+/-)) mouse phenotype is caused by mutations of the ribosomal protein L24 (Rpl24). Among various phenotypes in Bst(+/-) mice, the most interesting are its retinal abnormalities, consisting of delayed closure of choroid fissures, decreased ganglion cells and subretinal vascularization. We further characterized the Bst(+/-) mouse and investigated the underlying molecular mechanisms to assess the feasibility of using this strain as a model for stem cell therapy of retinal degenerative diseases due to retinal ganglion cell (RGC) loss. We found that, although RGCs are significantly reduced in retinal ganglion cell layer in Bst(+/-) mouse, melanopsin(+) RGCs, also called ipRGCs, appear to be unchanged. Pupillary light reflex was completely absent in Bst(+/-) mice but they had a normal circadian rhythm. In order to examine the pathological abnormalities in Bst(+/-) mice, we performed electron microscopy in RGC and found that mitochondria morphology was deformed, having irregular borders and lacking cristae. The complex activities of the mitochondrial electron transport chain were significantly decreased. Finally, for subretinal vascularization, we also found that angiogenesis is delayed in Bst(+/-) associated with delayed hyaloid regression. Characterization of Bst(+/-) retina suggests that the Bst(+/-) mouse strain could be a useful murine model. It might be used to explore further the pathogenesis and strategy of treatment of retinal degenerative diseases by employing stem cell technology.


Assuntos
Retina/patologia , Retina/fisiopatologia , Animais , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Mitocôndrias/metabolismo , Neovascularização Fisiológica , Consumo de Oxigênio , Fenótipo , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Opsinas de Bastonetes/metabolismo , Fator de Transcrição Brn-3A/metabolismo
20.
Am J Pathol ; 185(8): 2194-205, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26100214

RESUMO

Activation of Wnt/ß-catenin signaling during liver regeneration (LR) after partial hepatectomy (PH) is observed in several species. However, how this pathway is turned off when hepatocyte proliferation is no longer required is unknown. We assessed LR in liver-specific knockouts of Wntless (Wls-LKO), a protein required for Wnt secretion from a cell. When subjected to PH, Wls-LKO showed prolongation of hepatocyte proliferation for up to 4 days compared with littermate controls. This coincided with increased ß-catenin-T-cell factor 4 interaction and cyclin-D1 expression. Wls-LKO showed decreased expression and secretion of inhibitory Wnt5a during LR. Wnt5a expression increased between 24 and 48 hours, and Frizzled-2 between 24 and 72 hours, after PH in normal mice. Treatment of primary mouse hepatocytes and liver tumor cells with Wnt5a led to a notable decrease in ß-catenin-T-cell factor activity, cyclin-D1 expression, and cell proliferation. Intriguingly, Wnt5a-LKO did not display any prolongation of LR because of compensation by other cells. In addition, Wnt5a-LKO hepatocytes failed to respond to exogenous Wnt5a treatment in culture because of a compensatory decrease in Frizzled-2 expression. In conclusion, we demonstrate Wnt5a to be, by default, a negative regulator of ß-catenin signaling and hepatocyte proliferation, both in vitro and in vivo. We also provide evidence that the Wnt5a/Frizzled-2 axis suppresses ß-catenin signaling in hepatocytes in an autocrine manner, thereby contributing to timely conclusion of the LR process.


Assuntos
Proliferação de Células/fisiologia , Hepatócitos/metabolismo , Regeneração Hepática/fisiologia , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Regeneração Hepática/efeitos dos fármacos , Camundongos , Camundongos Knockout , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt-5a
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA