Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 599
Filtrar
1.
J Neurosci Res ; 102(7): e25361, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39034899

RESUMEN

Central and peripheral nervous system (CNS/PNS) proteoglycans (PGs) have diverse functional roles, this study examined how these control cellular behavior and tissue function. The CNS/PNS extracellular matrix (ECM) is a dynamic, responsive, highly interactive, space-filling, cell supportive, stabilizing structure maintaining tissue compartments, ionic microenvironments, and microgradients that regulate neuronal activity and maintain the neuron in an optimal ionic microenvironment. The CNS/PNS contains a high glycosaminoglycan content (60% hyaluronan, HA) and a diverse range of stabilizing PGs. Immobilization of HA in brain tissues by HA interactive hyalectan PGs preserves tissue hydration and neuronal activity, a paucity of HA in brain tissues results in a pro-convulsant epileptic phenotype. Diverse CS, KS, and HSPGs stabilize the blood-brain barrier and neurovascular unit, provide smart gel neurotransmitter neuron vesicle storage and delivery, organize the neuromuscular junction basement membrane, and provide motor neuron synaptic plasticity, and photoreceptor and neuron synaptic functions. PG-HA networks maintain ionic fluxes and microgradients and tissue compartments that contribute to membrane polarization dynamics essential to neuronal activation and neurotransduction. Hyalectans form neuroprotective perineuronal nets contributing to synaptic plasticity, memory, and cognitive learning. Sialoglycoprotein associated with cones and rods (SPACRCAN), an HA binding CSPG, stabilizes the inter-photoreceptor ECM. HSPGs pikachurin and eyes shut stabilize the photoreceptor synapse aiding in phototransduction and neurotransduction with retinal bipolar neurons crucial to visual acuity. This is achieved through Laminin G motifs in pikachurin, eyes shut, and neurexins that interact with the dystroglycan-cytoskeleton-ECM-stabilizing synaptic interconnections, neuronal interactive specificity, and co-ordination of regulatory action potentials in neural networks.


Asunto(s)
Astrocitos , Neuronas , Proteoglicanos , Animales , Proteoglicanos/metabolismo , Neuronas/metabolismo , Astrocitos/metabolismo , Matriz Extracelular/metabolismo , Humanos , Microambiente Celular/fisiología , Sistema Nervioso Central/metabolismo , Plasticidad Neuronal/fisiología
2.
Biomater Adv ; 163: 213952, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38991495

RESUMEN

Despite of being in different microenvironment, breast cancer cells influence the bone cells and persuade cancer metastasis from breast to bone. Multiple co-culture approaches have been explored to study paracrine signaling between these cells and to study the progression of cancer. However, lack of native tissue microenvironment remains a major bottleneck in existing co-culture technologies. Therefore, in the present study, a tumorigenic and an osteogenic microenvironment have been sutured together to create a multi-cellular environment and has been appraised to study cancer progression in bone tissue. The PCL-polystyrene and PCL-collagen fibrous scaffolds were characterized for tumorigenic and osteogenic potential induction on MDA-MB-231 and MC3T3-E1 cells respectively. Diffusion ability of crystal violet, glucose, and bovine serum albumin across the membrane were used to access the potential paracrine interaction facilitated by device. While in co-cultured condition, MDA-MB-231 cells showed EMT phenotype along with secretion of TNFα and PTHrP which lower down the expression of osteogenic markers including alkaline phosphatase, RUNX2, Osteocalcin and Osteoprotegerin. The cancer progression in bone microenvironment demonstrated the role and necessity of creating multiple tissue microenvironment and its contribution in studying multicellular disease progression and therapeutics.


Asunto(s)
Técnicas de Cocultivo , Osteogénesis , Humanos , Animales , Ratones , Osteogénesis/fisiología , Línea Celular Tumoral , Andamios del Tejido/química , Microambiente Tumoral/fisiología , Microambiente Celular/fisiología , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Femenino , Factor de Necrosis Tumoral alfa/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Comunicación Paracrina
3.
Neuroscience ; 553: 98-109, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-38964450

RESUMEN

Neurological disorders are a diverse group of conditions that can significantly impact individuals' quality of life. The maintenance of neural microenvironment homeostasis is essential for optimal physiological cellular processes. Perturbations in this delicate balance underlie various pathological manifestations observed across various neurological disorders. Current treatments for neurological disorders face substantial challenges, primarily due to the formidable blood-brain barrier and the intricate nature of neural tissue structures. These obstacles have resulted in a paucity of effective therapies and inefficiencies in patient care. Exosomes, nanoscale vesicles that contain a complex repertoire of biomolecules, are identifiable in various bodily fluids. They hold substantial promise in numerous therapeutic interventions due to their unique attributes, including targeted drug delivery mechanisms and the ability to cross the BBB, thereby enhancing their therapeutic potential. In this review, we investigate the therapeutic potential of exosomes across a range of neurological disorders, including neurodegenerative disorders, traumatic brain injury, peripheral nerve injury, brain tumors, and stroke. Through both in vitro and in vivo studies, our findings underscore the beneficial influence of exosomes in enhancing the neural microenvironment following neurological diseases, offering promise for improved neural recovery and management in these conditions.


Asunto(s)
Exosomas , Enfermedades del Sistema Nervioso , Exosomas/metabolismo , Humanos , Animales , Enfermedades del Sistema Nervioso/terapia , Enfermedades del Sistema Nervioso/metabolismo , Barrera Hematoencefálica/metabolismo , Microambiente Celular/fisiología
4.
Redox Rep ; 29(1): 2377870, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39010730

RESUMEN

OBJECTIVES: To observe the CISD2 expression among PCOS patients and to explore its profound impact on the follicular microenvironment. Moreover, we want to elucidate the intricate mechanistic contribution of CISD2 to the onset and progression of PCOS. METHODS: Oxidase NOX2, mitophagy-related proteins, and CISD2 were detected by WB. The changes in mitochondrial structure and quantity were observed by transmission electron microscopy. Mitochondrial and lysosome colocalization was used to detect the changes of mitophagy. MDA kit, GSH and GSSG Assay kit and ROS probe were used to detect oxidative stress damage. RESULTS: We found that CISD2, mitophagy and oxidase in the GCs of PCOS patients were significantly increased. Testosterone stimulation leads to the increase of oxidase, mitophagy, and CISD2 in KGN cells. CISD2 inhibition promoted the increase of mitophagy, and the activation of mitochondria-lysosome binding, while alleviating the oxidative stress. CONCLUSIONS: Inhibition of CISD2 can improve the occurrence of oxidative stress by increasing the level of mitophagy, thus affecting the occurrence and development of PCOS diseases.


Asunto(s)
Mitofagia , Estrés Oxidativo , Síndrome del Ovario Poliquístico , Adulto , Femenino , Humanos , Microambiente Celular/fisiología , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Mitofagia/efectos de los fármacos , Mitofagia/fisiología , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología
5.
Comput Biol Med ; 179: 108831, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38970834

RESUMEN

This work presents an advanced agent-based model developed within the FLAMEGPU2 framework, aimed at simulating the intricate dynamics of cell microenvironments. Our primary objective is to showcase FLAMEGPU2's potential in modelling critical features such as cell-cell and cell-ECM interactions, species diffusion, vascularisation, cell migration, and/or cell cycling. By doing so, we provide a versatile template that serves as a foundational platform for researchers to model specific biological mechanisms or processes. We highlight the utility of our approach as a microscale component within multiscale frameworks. Through four example applications, we demonstrate the model's versatility in capturing phenomena such as strain-stiffening behaviour of hydrogels, cell migration patterns within hydrogels, spheroid formation and fibre reorientation, and the simulation of diffusion processes within a vascularised and deformable domain. This work aims to bridge the gap between computational efficiency and biological fidelity, offering a scalable and flexible platform to advance our understanding of tissue biology and engineering.


Asunto(s)
Microambiente Celular , Simulación por Computador , Modelos Biológicos , Humanos , Microambiente Celular/fisiología , Movimiento Celular/fisiología , Hidrogeles/química
6.
Exp Neurol ; 379: 114867, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38914274

RESUMEN

An ischemic stroke (IS) is caused due to the lack of blood flow to cerebral tissue. Most of the studies have focused on how stroke affects the localized tissue, but it has been observed that a stroke can cause secondary complications in distant organs, such as Bone Marrow (BM). Our study focused on the effect of ischemic strokes on the bone marrow microenvironment. Bone marrow (BM) is a vital organ that maintains inflammatory homeostasis and aids in the repair of damaged tissue after injury/IS. We used the middle cerebral artery occlusion (MCAO) model of ischemic stroke on adult mice (6 months) and investigated the changes in the BM environment. BM cells were used for western blot and RT-PCR, and the BM supernatant was used for cytokine analysis and extracellular vesicle (EVs) isolation. We observed a significant increase in the total cell number within the BM and an increase in TNF-alpha and MCP-1, which are known for inducing a pro-inflammatory environment. Western blots analysis on the whole BM cell lysate demonstrated elevated levels of inflammatory factors (IL-6, TNF-alpha, and TLR-4) and senescence markers (p21 p16). EVs isolated from the BM supernatant showed no change in size or concentration; however, we found that the EVs carried increased miRNA-141-3p and miRNA-34a. Proteomic analysis on BM-derived EVs showed an alteration in the protein cargo of IS. We observed an increase in FgB, C3, Fn1, and Tra2b levels. The signaling pathway analysis showed mitochondrial function is most affected within the bone marrow. Our study demonstrated that IS induces changes in the BM environment and EVs secreted in the BM.


Asunto(s)
Médula Ósea , Vesículas Extracelulares , Accidente Cerebrovascular Isquémico , Ratones Endogámicos C57BL , Animales , Vesículas Extracelulares/metabolismo , Ratones , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Masculino , Médula Ósea/patología , Médula Ósea/metabolismo , Microambiente Celular/fisiología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Inflamación/metabolismo , Inflamación/patología , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/metabolismo , Citocinas/metabolismo
7.
Prog Retin Eye Res ; 102: 101275, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38797320

RESUMEN

The cornea is an ideal testing field for cell therapies. Its highly ordered structure, where specific cell populations are sequestered in different layers, together with its accessibility, has allowed the development of the first stem cell-based therapy approved by the European Medicine Agency. Today, different techniques have been proposed for autologous and allogeneic limbal and non-limbal cell transplantation. Cell replacement has also been attempted in cases of endothelial cell decompensation as it occurs in Fuchs dystrophy: injection of cultivated allogeneic endothelial cells is now in advanced phases of clinical development. Recently, stromal substitutes have been developed with excellent integration capability and transparency. Finally, cell-derived products, such as exosomes obtained from different sources, have been investigated for the treatment of severe corneal diseases with encouraging results. Optimization of the success rate of cell therapies obviously requires high-quality cultured cells/products, but the role of the surrounding microenvironment is equally important to allow engraftment of transplanted cells, to preserve their functions and, ultimately, lead to restoration of tissue integrity and transparency of the cornea.


Asunto(s)
Enfermedades de la Córnea , Trasplante de Células Madre , Humanos , Enfermedades de la Córnea/terapia , Trasplante de Células Madre/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Microambiente Celular/fisiología , Córnea , Trasplante de Córnea/métodos
8.
Adv Healthc Mater ; 13(21): e2400357, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38695274

RESUMEN

Multicellular organisms exhibit synergistic effects among their components, giving rise to emergent properties crucial for their genesis and overall functionality and survival. Morphogenesis involves and relies upon intricate and biunivocal interactions among cells and their environment, that is, the extracellular matrix (ECM). Cells secrete their own ECM, which in turn, regulates their morphogenetic program by controlling time and space presentation of matricellular signals. The ECM, once considered passive, is now recognized as an informative space where both biochemical and biophysical signals are tightly orchestrated. Replicating this sophisticated and highly interconnected informative media in a synthetic scaffold for tissue engineering is unattainable with current technology and this limits the capability to engineer functional human organs in vitro and in vivo. This review explores current limitations to in vitro organ morphogenesis, emphasizing the interplay of gene regulatory networks, mechanical factors, and tissue microenvironment cues. In vitro efforts to replicate biological processes for barrier organs such as the lung and intestine, are examined. The importance of maintaining cells within their native microenvironmental context is highlighted to accurately replicate organ-specific properties. The review underscores the necessity for microphysiological systems that faithfully reproduce cell-native interactions, for advancing the understanding of developmental disorders and disease progression.


Asunto(s)
Matriz Extracelular , Ingeniería de Tejidos , Humanos , Ingeniería de Tejidos/métodos , Matriz Extracelular/metabolismo , Animales , Microambiente Celular/fisiología , Andamios del Tejido/química , Pulmón/citología , Pulmón/metabolismo , Pulmón/fisiología
9.
Curr Osteoporos Rep ; 22(4): 416-432, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38782850

RESUMEN

PURPOSE OF THE REVIEW: In this review, we discuss the most recent scientific advances on the reciprocal regulatory interactions between the skeletal and hematopoietic stem cell niche, focusing on immunomodulation and its interplay with the cell's mitochondrial function, and how this impacts osteoimmune health during aging and disease. RECENT FINDINGS: Osteoimmunology investigates interactions between cells that make up the skeletal stem cell niche and immune system. Much work has investigated the complexity of the bone marrow microenvironment with respect to the skeletal and hematopoietic stem cells that regulate skeletal formation and immune health respectively. It has now become clear that these cellular components cooperate to maintain homeostasis and that dysfunction in their interaction can lead to aging and disease. Having a deeper, mechanistic appreciation for osteoimmune regulation will lead to better research perspective and therapeutics with the potential to improve the aging process, skeletal and hematologic regeneration, and disease targeting.


Asunto(s)
Envejecimiento , Médula Ósea , Células Madre Hematopoyéticas , Homeostasis , Nicho de Células Madre , Humanos , Envejecimiento/fisiología , Envejecimiento/inmunología , Médula Ósea/inmunología , Nicho de Células Madre/fisiología , Huesos/metabolismo , Huesos/inmunología , Mitocondrias , Microambiente Celular/fisiología , Células de la Médula Ósea/inmunología , Animales , Inmunomodulación
10.
Mucosal Immunol ; 17(1): 41-53, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37866719

RESUMEN

Despite their importance for immunity against sexually transmitted infections, the composition of female reproductive tract (FRT) memory T-cell populations in response to changes within the local tissue environment under the regulation of the menstrual cycle remains poorly defined. Here, we show that in humans and pig-tailed macaques, the cycle determines distinct clusters of differentiation 4 T-cell surveillance behaviors by subsets corresponding to migratory memory (TMM) and resident memory T cells. TMM displays tissue-itinerant trafficking characteristics, restricted distribution within the FRT microenvironment, and distinct effector responses to infection. Gene pathway analysis by RNA sequencing identified TMM-specific enrichment of genes involved in hormonal regulation and inflammatory responses. FRT T-cell subset fluctuations were discovered that synchronized to cycle-driven CCR5 signaling. Notably, oral administration of a CCR5 antagonist drug blocked TMM trafficking. Taken together, this study provides novel insights into the dynamic nature of FRT memory CD4 T cells and identifies the menstrual cycle as a key regulator of immune surveillance at the site of STI pathogen exposure.


Asunto(s)
Linfocitos T CD4-Positivos , Genitales Femeninos , Ciclo Menstrual , Receptores CCR5 , Transducción de Señal , Femenino , Humanos , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Genitales Femeninos/inmunología , Genitales Femeninos/metabolismo , Ciclo Menstrual/inmunología , Ciclo Menstrual/fisiología , Receptores CCR5/genética , Receptores CCR5/metabolismo , Subgrupos de Linfocitos T/inmunología , Macaca nemestrina/inmunología , Memoria Inmunológica , Microambiente Celular/inmunología , Microambiente Celular/fisiología , Antagonistas de los Receptores CCR5/farmacología
11.
Adv Sci (Weinh) ; 10(2): e2203308, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36424137

RESUMEN

Diabetic foot ulcers (DFU), one of the most serious complications of diabetes, are essentially chronic, nonhealing wounds caused by diabetic neuropathy, vascular disease, and bacterial infection. Given its pathogenesis, the DFU microenvironment is rather complicated and characterized by hyperglycemia, ischemia, hypoxia, hyperinflammation, and persistent infection. However, the current clinical therapies for DFU are dissatisfactory, which drives researchers to turn attention to advanced nanotechnology to address DFU therapeutic bottlenecks. In the last decade, a large number of multifunctional nanosystems based on the microenvironment of DFU have been developed with positive effects in DFU therapy, forming a novel concept of "DFU nanomedicine". However, a systematic overview of DFU nanomedicine is still unavailable in the literature. This review summarizes the microenvironmental characteristics of DFU, presents the main progress of wound healing, and summaries the state-of-the-art therapeutic strategies for DFU. Furthermore, the main challenges and future perspectives in this field are discussed and prospected, aiming to fuel and foster the development of DFU nanomedicines successfully.


Asunto(s)
Microambiente Celular , Pie Diabético , Nanomedicina , Humanos , Diabetes Mellitus , Pie Diabético/tratamiento farmacológico , Pie Diabético/fisiopatología , Hiperglucemia , Cicatrización de Heridas , Microambiente Celular/fisiología
12.
Int J Mol Sci ; 23(4)2022 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-35216117

RESUMEN

Stem cells (SC) are largely known for their potential to restore damaged tissue through various known mechanisms. Among these mechanisms is their ability to transfer healthy mitochondria to injured cells to rescue them. This mitochondrial transfer plays a critical role in the healing process. To determine the optimal parameters for inducing mitochondrial transfer between cells, we assessed mitochondrial transfer as a function of seeding density and in two-dimensional (2D) and semi three-dimensional (2.5D) culture models. Since mitochondrial transfer can occur through direct contact or secretion, the 2.5D culture model utilizes collagen to provide cells with a more physiologically relevant extracellular matrix and offers a more realistic representation of cell attachment and movement. Results demonstrate the dependence of mitochondrial transfer on cell density and the distance between donor and recipient cell. Furthermore, the differences found between the transfer of mitochondria in 2D and 2.5D microenvironments suggest an optimal mode of mitochondria transport. Using these parameters, we explored the effects on mitochondrial transfer between SCs and tumorigenic cells. HEK293 (HEK) is an immortalized cell line derived from human embryonic kidney cells which grow rapidly and form tumors in culture. Consequently, HEKs have been deemed tumorigenic and are widely used in cancer research. We observed mitochondrial transfer from SCs to HEK cells at significantly higher transfer rates when compared to a SC-SC co-culture system. Interestingly, our results also revealed an increase in the migratory ability of HEK cells when cultured with SCs. As more researchers find co-localization of stem cells and tumors in the human body, these results could be used to better understand their biological relationship and lead to enhanced therapeutic applications.


Asunto(s)
Tejido Adiposo/fisiología , Microambiente Celular/fisiología , Células Madre Mesenquimatosas/fisiología , Mitocondrias/fisiología , Adipocitos/fisiología , Carcinogénesis/patología , Recuento de Células/métodos , Línea Celular , Técnicas de Cocultivo/métodos , Células HEK293 , Humanos
13.
J Immunol ; 208(5): 1292-1304, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35131868

RESUMEN

Pathogen-specific CD8 T cells face the problem of finding rare cells that present their cognate Ag either in the lymph node or in infected tissue. Although quantitative details of T cell movement strategies in some tissues such as lymph nodes or skin have been relatively well characterized, we still lack quantitative understanding of T cell movement in many other important tissues, such as the spleen, lung, liver, and gut. We developed a protocol to generate stable numbers of liver-located CD8 T cells, used intravital microscopy to record movement patterns of CD8 T cells in livers of live mice, and analyzed these and previously published data using well-established statistical and computational methods. We show that, in most of our experiments, Plasmodium-specific liver-localized CD8 T cells perform correlated random walks characterized by transiently superdiffusive displacement with persistence times of 10-15 min that exceed those observed for T cells in lymph nodes. Liver-localized CD8 T cells typically crawl on the luminal side of liver sinusoids (i.e., are in the blood); simulating T cell movement in digital structures derived from the liver sinusoids illustrates that liver structure alone is sufficient to explain the relatively long superdiffusive displacement of T cells. In experiments when CD8 T cells in the liver poorly attach to the sinusoids (e.g., 1 wk after immunization with radiation-attenuated Plasmodium sporozoites), T cells also undergo Lévy flights: large displacements occurring due to cells detaching from the endothelium, floating with the blood flow, and reattaching at another location. Our analysis thus provides quantitative details of movement patterns of liver-localized CD8 T cells and illustrates how structural and physiological details of the tissue may impact T cell movement patterns.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Movimiento Celular/fisiología , Hígado/inmunología , Malaria/prevención & control , Plasmodium berghei/inmunología , Animales , Capilares/citología , Microambiente Celular/fisiología , Hígado/irrigación sanguínea , Malaria/patología , Ratones , Plasmodium berghei/crecimiento & desarrollo , Esporozoítos/crecimiento & desarrollo , Esporozoítos/inmunología , Vacunación
14.
PLoS One ; 17(1): e0262532, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35085314

RESUMEN

This study aimed to report the effects of different doses of ionizing radiation on inflammatory and repair stage of human skin graft adherence in Nude mice wounds. Animals were divided into transplanted with irradiated human skin grafts (IHSG) at 25 and 50 kGy (IHSG 25 kGy; IHSG 50 kGy) and non-IHSG and euthanized on the 3rd, 7th and 21st days after the surgery, by gross and microscopic changes, immunostaining for human type I collagen (Col I) and mouse Col I and Col III and inflammatory cells. We found an effectiveness of human split-thickness graft adherence in mice transplanted with IHSG 25 kGy, as well decrease in dermo-epidermal necrosis and neutrophils, lower loss of skin thickness, epithelization and neo-vascularization. Day 21 post-transplantation with IHSG 25 kGy was observed a well-preserved human skin in the border of the graft, a prominent granulation tissue in an organization by proliferated fibroblasts, Col III deposition and increased B-cells and macrophages. A complete adherence of human skin graft occurred with IHSG 25 kGy. We suggest that the ionizing radiation at 25 kGy mediates inflammation and the repair stage of human skin graft adherence in murine model, thus emerging as a potential tool in healing cutaneous wounds.


Asunto(s)
Microambiente Celular/fisiología , Colágeno Tipo I/metabolismo , Piel/metabolismo , Piel/fisiopatología , Adherencias Tisulares/metabolismo , Adherencias Tisulares/fisiopatología , Cicatrización de Heridas/fisiología , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Repitelización/fisiología , Trasplante de Piel/métodos , Piel Artificial
15.
Lipids Health Dis ; 21(1): 5, 2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-34996476

RESUMEN

Recent advances in society have resulted in the emergence of both hyperlipidemia and obesity as life-threatening conditions in people with implications for various types of diseases, such as cardiovascular diseases and cancer. This is further complicated by a global rise in the aging population, especially menopausal women, who mostly suffer from overweight and bone loss simultaneously. Interestingly, clinical observations in these women suggest that osteoarthritis may be linked to a higher body mass index (BMI), which has led many to believe that there may be some degree of bone dysfunction associated with conditions such as obesity. It is also common practice in many outpatient settings to encourage patients to control their BMI and lose weight in an attempt to mitigate mechanical stress and thus reduce bone pain and joint dysfunction. Together, studies show that bone is not only a mechanical organ but also a critical component of metabolism, and various endocrine functions, such as calcium metabolism. Numerous studies have demonstrated a relationship between metabolic dysfunction in bone and abnormal lipid metabolism. Previous studies have also regarded obesity as a metabolic disorder. However, the relationship between lipid metabolism and bone metabolism has not been fully elucidated. In this narrative review, the data describing the close relationship between bone and lipid metabolism was summarized and the impact on both the normal physiology and pathophysiology of these tissues was discussed at both the molecular and cellular levels.


Asunto(s)
Huesos/metabolismo , Metabolismo de los Lípidos , Animales , Enfermedades Óseas/metabolismo , Enfermedades Óseas/fisiopatología , Neoplasias Óseas/metabolismo , Neoplasias Óseas/fisiopatología , Huesos/fisiología , Huesos/fisiopatología , Microambiente Celular/fisiología , Colesterol/metabolismo , Colesterol/fisiología , Humanos , Metabolismo de los Lípidos/fisiología , Osteoporosis/metabolismo
16.
Diabetes ; 71(1): 23-30, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34995348

RESUMEN

Age-related immunosenescence, defined as an increase in inflammaging and the decline of the immune system, leads to tissue dysfunction and increased risk for metabolic disease. The elderly population is expanding, leading to a heightened need for therapeutics to improve health span. With age, many alterations of the immune system are observed, including shifts in the tissue-resident immune cells, increased expression of inflammatory factors, and the accumulation of senescent cells, all of which are responsible for a chronic inflammatory loop. Adipose tissue and the immune cell activation within are of particular interest for their well-known roles in metabolic disease. Recent literature reveals that adipose tissue is an organ in which signs of initial aging occur, including immune cell activation. Aged adipose tissue reveals changes in many innate and adaptive immune cell subsets, revealing a complex interaction that contributes to inflammation, increased senescence, impaired catecholamine-induced lipolysis, and impaired insulin sensitivity. Here, we will describe current knowledge surrounding age-related changes in immune cells while relating those findings to recent discoveries regarding immune cells in aged adipose tissue.


Asunto(s)
Tejido Adiposo/patología , Microambiente Celular/fisiología , Senescencia Celular/fisiología , Inflamación/patología , Leucocitos/fisiología , Tejido Adiposo/metabolismo , Anciano , Envejecimiento/sangre , Envejecimiento/inmunología , Envejecimiento/metabolismo , Animales , Microambiente Celular/inmunología , Femenino , Humanos , Inmunosenescencia/fisiología , Inflamación/metabolismo , Macrófagos/fisiología , Masculino , Ratones
17.
JCI Insight ; 7(1)2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34793332

RESUMEN

Alport syndrome (AS) is a genetic disorder caused by mutations in type IV collagen that lead to defective glomerular basement membrane, glomerular filtration barrier (GFB) damage, and progressive chronic kidney disease. While the genetic basis of AS is well known, the molecular and cellular mechanistic details of disease pathogenesis have been elusive, hindering the development of mechanism-based therapies. Here, we performed intravital multiphoton imaging of the local kidney tissue microenvironment in a X-linked AS mouse model to directly visualize the major drivers of AS pathology. Severely distended glomerular capillaries and aneurysms were found accompanied by numerous microthrombi, increased glomerular endothelial surface layer (glycocalyx) and immune cell homing, GFB albumin leakage, glomerulosclerosis, and interstitial fibrosis by 5 months of age, with an intermediate phenotype at 2 months. Renal histology in mouse or patient tissues largely failed to detect capillary aberrations. Treatment of AS mice with hyaluronidase or the ACE inhibitor enalapril reduced the excess glomerular endothelial glycocalyx and blocked immune cell homing and GFB albumin leakage. This study identified central roles of glomerular mechanical forces and endothelial and immune cell activation early in AS, which could be therapeutically targeted to reduce mechanical strain and local tissue inflammation and improve kidney function.


Asunto(s)
Capilares , Microscopía Intravital , Glomérulos Renales , Nefritis Hereditaria , Animales , Capilares/diagnóstico por imagen , Capilares/inmunología , Capilares/patología , Microambiente Celular/fisiología , Modelos Animales de Enfermedad , Humanos , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/diagnóstico por imagen , Glomérulos Renales/inmunología , Glomérulos Renales/patología , Masculino , Ratones , Nefritis Hereditaria/diagnóstico por imagen , Nefritis Hereditaria/patología
18.
Andrology ; 10(1): 92-104, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34420258

RESUMEN

BACKGROUND: Terminally differentiated mammalian sperm are exposed to gradients of viscosity, pH, and osmolality both in the male and female reproductive tract during their perilous journey to quest the ovum. The complex physicochemical factors play an integral role in preparing sperm for the fertilization process. OBJECTIVES: To elucidate the influence of the reproductive tract microenvironment especially viscosity, pH, and osmolality in regulating sperm functional and fertilization competence. MATERIALS AND METHODS: The data used in this review were collected from the research papers and online databases focusing on the influence of viscosity, pH, and osmolality on sperm function. DISCUSSION: The gradients of viscosity, pH, and osmolality exist across various segments of the male and female reproductive tract. The changes in the viscosity create a physical barrier, pH aid in capacitation and hyperactivation, and the osmotic stress selects a progressive sperm subpopulation for accomplishing fertilization. The sperm function tests are developed based on the concept that the male genotype is the major contributor to the reproductive outcome. However, recent studies demonstrate the significance of sperm genotype-environment interactions that are essentially contributing to reproductive success. Hence, it is imperative to assess the impact of physicochemical stresses and the adaptive ability of the terminally differentiated sperm, which in turn would improve the outcome of the assisted reproductive technologies and male fertility assessment. CONCLUSION: Elucidating the influence of the reproductive tract microenvironment on sperm function provides newer insights into the procedures that need to be adopted for selecting fertile males for breeding, and ejaculates for the assisted reproductive technologies.


Asunto(s)
Microambiente Celular/fisiología , Fertilidad/fisiología , Genitales/citología , Espermatozoides/química , Animales , Femenino , Humanos , Concentración de Iones de Hidrógeno , Masculino , Concentración Osmolar , Interacciones Espermatozoide-Óvulo/fisiología , Viscosidad
19.
Front Immunol ; 12: 768439, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858426

RESUMEN

The shape and spatial organization -the anatomy- of a tissue profoundly influences its function. Knowledge of the anatomical relationships between parent and daughter cells is necessary to understand differentiation and how the crosstalk between the different cells in the tissue leads to physiological maintenance and pathological perturbations. Blood cell production takes place in the bone marrow through the progressive differentiation of stem cells and progenitors. These are maintained and regulated by a heterogeneous microenvironment composed of stromal and hematopoietic cells. While hematopoiesis has been studied in extraordinary detail through functional and multiomics approaches, much less is known about the spatial organization of blood production and how local cues from the microenvironment influence this anatomy. Here, we discuss some of the studies that revealed a complex anatomy of hematopoiesis where discrete local microenvironments spatially organize and regulate specific subsets of hematopoietic stem cells and/or progenitors. We focus on the open questions in the field and discuss how new tools and technological advances are poised to transform our understanding of the anatomy of hematopoiesis.


Asunto(s)
Células de la Médula Ósea/fisiología , Médula Ósea/fisiología , Microambiente Celular/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Nicho de Células Madre/fisiología , Animales , Médula Ósea/anatomía & histología , Células de la Médula Ósea/citología , Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/citología , Humanos , Modelos Biológicos
20.
Front Immunol ; 12: 757827, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925328

RESUMEN

Kingella kingae is an emerging pathogen that causes septic arthritis, osteomyelitis, and bacteremia in children from 6 to 48 months of age. The presence of bacteria within or near the bone is associated with an inflammatory process that results in osteolysis, but the underlying pathogenic mechanisms involved are largely unknown. To determine the link between K. kingae and bone loss, we have assessed whether infection per se or through the genesis of a pro-inflammatory microenvironment can promote osteoclastogenesis. For that purpose, we examined both the direct effect of K. kingae and the immune-mediated mechanism involved in K. kingae-infected macrophage-induced osteoclastogenesis. Our results indicate that osteoclastogenesis is stimulated by K. kingae infection directly and indirectly by fueling a potent pro-inflammatory response that drives macrophages to undergo functional osteoclasts via TNF-α and IL-1ß induction. Such osteoclastogenic capability of K. kingae is counteracted by their outer membrane vesicles (OMV) in a concentration-dependent manner. In conclusion, this model allowed elucidating the interplay between the K. kingae and their OMV to modulate osteoclastogenesis from exposed macrophages, thus contributing to the modulation in joint and bone damage.


Asunto(s)
Microambiente Celular/fisiología , Infecciones por Neisseriaceae/inmunología , Infecciones por Neisseriaceae/patología , Osteoclastos/inmunología , Osteogénesis/fisiología , Animales , Línea Celular , Humanos , Kingella kingae , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Osteoclastos/metabolismo , Células RAW 264.7
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...