Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 598
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Rev Mol Cell Biol ; 19(11): 731-745, 2018 11.
Article in English | MEDLINE | ID: mdl-30305710

ABSTRACT

Mammalian cells respond to stress by activating mechanisms that support cellular functions and hence maintain microenvironmental and organismal homeostasis. Intracellular responses to stress, their regulation and their pathophysiological implications have been extensively studied. However, little is known about the signals that emanate from stressed cells to enable a coordinated adaptive response across tissues, organs and the whole organism. Considerable evidence has now accumulated indicating that the intracellular mechanisms that are activated in response to different stresses - which include the DNA damage response, the unfolded protein response, mitochondrial stress signalling and autophagy - as well as the mechanisms ensuring the proliferative inactivation or elimination of terminally damaged cells - such as cell senescence and regulated cell death - are all coupled with the generation of signals that elicit microenvironmental and/or systemic responses. These signals, which involve changes in the surface of stressed cells and/or the secretion of soluble factors or microvesicles, generally support systemic homeostasis but can also contribute to maladaptation and disease.


Subject(s)
Homeostasis/physiology , Stress, Physiological/physiology , Animals , Cellular Microenvironment/physiology , Cellular Senescence/physiology , Humans , Signal Transduction/physiology
2.
Nat Rev Mol Cell Biol ; 18(12): 743-757, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29115298

ABSTRACT

The way in which cells coordinate their behaviours during various biological processes, including morphogenesis, cancer progression and tissue remodelling, largely depends on the mechanical properties of the external environment. In contrast to single cells, collective cell behaviours rely on the cellular interactions not only with the surrounding extracellular matrix but also with neighbouring cells. Collective dynamics is not simply the result of many individually moving blocks. Instead, cells coordinate their movements by actively interacting with each other. These mechanisms are governed by mechanosensitive adhesion complexes at the cell-substrate interface and cell-cell junctions, which respond to but also further transmit physical signals. The mechanosensitivity and mechanotransduction at adhesion complexes are important for regulating tissue cohesiveness and thus are important for collective cell behaviours. Recent studies have shown that the physical properties of the cellular environment, which include matrix stiffness, topography, geometry and the application of external forces, can alter collective cell behaviours, tissue organization and cell-generated forces. On the basis of these findings, we can now start building our understanding of the mechanobiology of collective cell movements that span over multiple length scales from the molecular to the tissue level.


Subject(s)
Cell Communication/physiology , Cell Movement/physiology , Cellular Microenvironment/physiology , Extracellular Matrix/physiology , Mechanotransduction, Cellular/physiology , Animals , Humans
3.
Genes Dev ; 33(3-4): 127-143, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30709901

ABSTRACT

Originally thought of as a stress response end point, the view of cellular senescence has since evolved into one encompassing a wide range of physiological and pathological functions, including both protumorignic and antitumorigenic features. It has also become evident that senescence is a highly dynamic and heterogenous process. Efforts to reconcile the beneficial and detrimental features of senescence suggest that physiological functions require the transient presence of senescent cells in the tissue microenvironment. Here, we propose the concept of a physiological "senescence life cycle," which has pathological consequences if not executed in its entirety.


Subject(s)
Cell Cycle/physiology , Cellular Senescence/physiology , Neoplasms/physiopathology , Cellular Microenvironment/physiology , Epigenomics , Humans , Precancerous Conditions/physiopathology , Telomere Shortening
4.
Nat Rev Mol Cell Biol ; 15(10): 647-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25237826

ABSTRACT

Mammalian organs are challenging to study as they are fairly inaccessible to experimental manipulation and optical observation. Recent advances in three-dimensional (3D) culture techniques, coupled with the ability to independently manipulate genetic and microenvironmental factors, have enabled the real-time study of mammalian tissues. These systems have been used to visualize the cellular basis of epithelial morphogenesis, to test the roles of specific genes in regulating cell behaviours within epithelial tissues and to elucidate the contribution of microenvironmental factors to normal and disease processes. Collectively, these novel models can be used to answer fundamental biological questions and generate replacement human tissues, and they enable testing of novel therapeutic approaches, often using patient-derived cells.


Subject(s)
Cell Culture Techniques , Epithelial Cells/physiology , Epithelium/growth & development , Morphogenesis/physiology , Animals , Cell Proliferation , Cellular Microenvironment/physiology , Epithelial Cells/cytology , Epithelium/embryology , Gene Expression Regulation , Mammals , Organ Culture Techniques
5.
J Neurosci Res ; 102(7): e25361, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39034899

ABSTRACT

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.


Subject(s)
Astrocytes , Neurons , Proteoglycans , Animals , Proteoglycans/metabolism , Neurons/metabolism , Astrocytes/metabolism , Extracellular Matrix/metabolism , Humans , Cellular Microenvironment/physiology , Central Nervous System/metabolism , Neuronal Plasticity/physiology
6.
J Immunol ; 208(5): 1292-1304, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35131868

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Movement/physiology , Liver/immunology , Malaria/prevention & control , Plasmodium berghei/immunology , Animals , Capillaries/cytology , Cellular Microenvironment/physiology , Liver/blood supply , Malaria/pathology , Mice , Plasmodium berghei/growth & development , Sporozoites/growth & development , Sporozoites/immunology , Vaccination
7.
Curr Osteoporos Rep ; 22(4): 416-432, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38782850

ABSTRACT

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.


Subject(s)
Aging , Bone Marrow , Hematopoietic Stem Cells , Homeostasis , Stem Cell Niche , Humans , Aging/physiology , Aging/immunology , Bone Marrow/immunology , Stem Cell Niche/physiology , Bone and Bones/metabolism , Bone and Bones/immunology , Mitochondria , Cellular Microenvironment/physiology , Bone Marrow Cells/immunology , Animals , Immunomodulation
8.
Proc Natl Acad Sci U S A ; 117(36): 22473-22483, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32848055

ABSTRACT

Human fungal infections may fail to respond to contemporary antifungal therapies in vivo despite in vitro fungal isolate drug susceptibility. Such a discrepancy between in vitro antimicrobial susceptibility and in vivo treatment outcomes is partially explained by microbes adopting a drug-resistant biofilm mode of growth during infection. The filamentous fungal pathogen Aspergillus fumigatus forms biofilms in vivo, and during biofilm growth it has reduced susceptibility to all three classes of contemporary antifungal drugs. Specific features of filamentous fungal biofilms that drive antifungal drug resistance remain largely unknown. In this study, we applied a fluorescence microscopy approach coupled with transcriptional bioreporters to define spatial and temporal oxygen gradients and single-cell metabolic activity within A. fumigatus biofilms. Oxygen gradients inevitably arise during A. fumigatus biofilm maturation and are both critical for, and the result of, A. fumigatus late-stage biofilm architecture. We observe that these self-induced hypoxic microenvironments not only contribute to filamentous fungal biofilm maturation but also drive resistance to antifungal treatment. Decreasing oxygen levels toward the base of A. fumigatus biofilms increases antifungal drug resistance. Our results define a previously unknown mechanistic link between filamentous fungal biofilm physiology and contemporary antifungal drug resistance. Moreover, we demonstrate that drug resistance mediated by dynamic oxygen gradients, found in many bacterial biofilms, also extends to the fungal kingdom. The conservation of hypoxic drug-resistant niches in bacterial and fungal biofilms is thus a promising target for improving antimicrobial therapy efficacy.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus fumigatus , Biofilms/drug effects , Cellular Microenvironment , Drug Resistance, Fungal , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/physiology , Cell Hypoxia , Cellular Microenvironment/drug effects , Cellular Microenvironment/physiology , Oxygen/pharmacology
9.
Development ; 146(13)2019 07 05.
Article in English | MEDLINE | ID: mdl-31278123

ABSTRACT

The process of cell competition results in the 'elimination of cells that are viable but less fit than surrounding cells'. Given the highly heterogeneous nature of our tissues, it seems increasingly likely that cells are engaged in a 'survival of the fittest' battle throughout life. The process has a myriad of positive roles in the organism: it selects against mutant cells in developing tissues, prevents the propagation of oncogenic cells and eliminates damaged cells during ageing. However, 'super-fit' cancer cells can exploit cell competition mechanisms to expand and spread. Here, we review the regulation, roles and risks of cell competition in organism development, ageing and disease.


Subject(s)
Cell Communication/physiology , Cell Physiological Phenomena , Competitive Behavior/physiology , Genetic Fitness/physiology , Selection, Genetic/physiology , Aging/physiology , Animals , Cell Physiological Phenomena/genetics , Cellular Microenvironment/physiology , Humans , Reproduction/physiology
10.
J Pathol ; 255(2): 141-154, 2021 10.
Article in English | MEDLINE | ID: mdl-34173975

ABSTRACT

Stromal-epithelial interactions are critical to the morphogenesis, differentiation, and homeostasis of the prostate, but the molecular identity and anatomy of discrete stromal cell types is poorly understood. Using single-cell RNA sequencing, we identified and validated the in situ localization of three smooth muscle subtypes (prostate smooth muscle, pericytes, and vascular smooth muscle) and two novel fibroblast subtypes in human prostate. Peri-epithelial fibroblasts (APOD+) wrap around epithelial structures, whereas interstitial fibroblasts (C7+) are interspersed in extracellular matrix. In contrast, the mouse displayed three fibroblast subtypes with distinct proximal-distal and lobe-specific distribution patterns. Statistical analysis of mouse and human fibroblasts showed transcriptional correlation between mouse prostate (C3+) and urethral (Lgr5+) fibroblasts and the human interstitial fibroblast subtype. Both urethral fibroblasts (Lgr5+) and ductal fibroblasts (Wnt2+) in the mouse contribute to a proximal Wnt/Tgfb signaling niche that is absent in human prostate. Instead, human peri-epithelial fibroblasts express secreted WNT inhibitors SFRPs and DKK1, which could serve as a buffer against stromal WNT ligands by creating a localized signaling niche around individual prostate glands. We also identified proximal-distal fibroblast density differences in human prostate that could amplify stromal signaling around proximal prostate ducts. In human benign prostatic hyperplasia, fibroblast subtypes upregulate critical immunoregulatory pathways and show distinct distributions in stromal and glandular phenotypes. A detailed taxonomy of leukocytes in benign prostatic hyperplasia reveals an influx of myeloid dendritic cells, T cells and B cells, resembling a mucosal inflammatory disorder. A receptor-ligand interaction analysis of all cell types revealed a central role for fibroblasts in growth factor, morphogen, and chemokine signaling to endothelia, epithelia, and leukocytes. These data are foundational to the development of new therapeutic targets in benign prostatic hyperplasia. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cellular Microenvironment/physiology , Fibroblasts/cytology , Prostate/cytology , Animals , Extracellular Matrix , Humans , Male , Mice , Prostatic Hyperplasia/pathology , Single-Cell Analysis
11.
J Immunol ; 204(12): 3262-3272, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32332108

ABSTRACT

The self-renewal ability is a unique property of fetal-derived innate-like B-1a lymphocytes, which survive and function without being replenished by bone marrow (BM) progenitors. However, the mechanism by which IgM-secreting mature B-1a lymphocytes self-renew is poorly understood. In this study, we showed that Bmi1 was critically involved in this process. Although Bmi1 is considered essential for lymphopoiesis, the number of mature conventional B cells was not altered when Bmi1 was deleted in the B cell lineage. In contrast, the number of peritoneal B-1a cells was significantly reduced. Peritoneal cell transfer assays revealed diminished self-renewal ability of Bmi1-deleted B-1a cells, which was restored by additional deletion of Ink4-Arf, the well-known target of Bmi1 Fetal liver cells with B cell-specific Bmi1 deletion failed to repopulate peritoneal B-1a cells, but not other B-2 lymphocytes after transplantation assays, suggesting that Bmi1 may be involved in the developmental process of B-1 progenitors to mature B-1a cells. Although Bmi1 deletion has also been shown to alter the microenvironment for hematopoietic stem cells, fat-associated lymphoid clusters, the reported niche for B-1a cells, were not impaired in Bmi1 -/- mice. RNA expression profiling suggested lysine demethylase 5B (Kdm5b) as another possible target of Bmi1, which was elevated in Bmi1-/- B-1a cells in a stress setting and might repress B-1a cell proliferation. Our work has indicated that Bmi1 plays pivotal roles in self-renewal and maintenance of fetal-derived B-1a cells.


Subject(s)
B-Lymphocyte Subsets/metabolism , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Animals , B-Lymphocyte Subsets/physiology , Bone Marrow/metabolism , Cell Lineage/physiology , Cell Proliferation/physiology , Cells, Cultured , Cellular Microenvironment/physiology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Lymphocytes/metabolism , Lymphocytes/physiology , Lymphopoiesis/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID
12.
J Immunol ; 205(11): 3179-3190, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33115854

ABSTRACT

Air pollution exposure leads to various inflammatory diseases in the human respiratory system. Chronic rhinosinusitis is an inflammatory disease caused by viruses, bacteria, or air pollutants. However, the underlying molecular mechanisms through which air particulate matter (PM) causes inflammation and disease remain unclear. In this article, we report that the induction of exosomal microRNAs (miRNAs) from human nasal epithelial cells upon airborne PM exposure promotes proinflammatory M1 macrophage polarization via downregulated RORα expression. Exposure of human nasal epithelial cells to PM results in inflammation-related miRNA expression, and more miRNA is secreted through exosomes delivered to macrophages. Among these, miRNA-19a and miRNA-614 directly bind to the 3'-untranslated region of RORα mRNA and downregulate RORα expression, which leads to inflammation due to inflammatory cytokine upregulation and induces macrophages to a proinflammatory M1-like state. Finally, we showed enhanced expression of miRNA-19a and miRNA-614 but reduced RORα expression in a chronic rhinosinusitis patient tissue compared with the normal. Altogether, our results suggest that PM-induced exosomal miRNAs might play a crucial role in the proinflammatory mucosal microenvironment and macrophage polarization through the regulation of RORα expression.


Subject(s)
Air Pollutants/adverse effects , Exosomes/metabolism , Inflammation/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Respiratory Mucosa/metabolism , Cell Line , Cellular Microenvironment/drug effects , Cellular Microenvironment/physiology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Exosomes/drug effects , Humans , Inflammation/chemically induced , Macrophages/drug effects , Particulate Matter/adverse effects , Respiratory Mucosa/drug effects , THP-1 Cells
13.
Exp Cell Res ; 398(1): 112398, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33271129

ABSTRACT

The term vascular niche indicate the physical and biochemical microenvironment around blood vessel where endothelial cells, pericytes, and smooth muscle cells organize themselves to form blood vessels and release molecules involved in the recruitment of hematopoietic stem cells, endothelial progenitor cells and mesenchymal stem cells. The vascular niche creates a permissive environment that enables different cell types to realize their developmental or regenerative programs. In this context, the proximity between the endothelium and the new-forming cellular components of organs suggests an essential role of endothelial cells in the organs maturation. Dynamic interactions between specific organ endothelial cells and different cellular conponents are crucial for different organ morphogenesis and function. Conversely, organs provide cues shaping vascular network structure.


Subject(s)
Endothelial Cells/metabolism , Endothelial Cells/physiology , Regeneration , Animals , Cellular Microenvironment/physiology , Endothelial Cells/cytology , Humans , Organogenesis
14.
Exp Cell Res ; 408(1): 112837, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34547255

ABSTRACT

Myelodysplastic syndrome (MDS) is a heterogeneous group of clonal hematopoietic neoplasms. The progression of malignancy is closely associated with immune regulation. Macrophages are indispensable tissue components and have been proposed to play a role in the pathophysiology of hematopoietic malignancies. However, the specific role of macrophages in the development of MDS remains unclear. Here, we investigated the characteristics and phenotypic evolution of macrophages from patients with MDS. Macrophages from patients with MDS expressed CD68, CD86 and CD163. Furthermore, MDS macrophages exhibited more M2-related characteristics. Moreover, a number of phenotype-associated genes in MDS macrophages exhibited diverse responses to iron overload or iron chelation upon stimulation by ferric chloride or deferoxamine (DFO, an iron chelator). Ferric chloride polarized MDS macrophages to exhibit more M1-related characteristics, a phenomenon that could be partially reversed by DFO. Therefore, this study reveals the characteristics and phenotypic evolution of MDS macrophages and broadens the knowledge of macrophage plasticity in hematopoietic malignancies.


Subject(s)
Iron Overload/pathology , Iron/metabolism , Macrophages/pathology , Myelodysplastic Syndromes/pathology , Adult , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cellular Microenvironment/physiology , Chlorides/metabolism , Female , Ferric Compounds/metabolism , Humans , Iron Overload/metabolism , Male , Middle Aged , Receptors, Cell Surface/metabolism
15.
Cell Mol Life Sci ; 78(23): 7161-7183, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34635950

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells (IMCs) with immunosuppressive functions, whereas IMCs originally differentiate into granulocytes, macrophages, and dendritic cells (DCs) to participate in innate immunity under steady-state conditions. At present, difficulties remain in identifying MDSCs due to lacking of specific biomarkers. To make identification of MDSCs accurately, it also needs to be determined whether having immunosuppressive functions. MDSCs play crucial roles in anti-tumor, angiogenesis, and metastasis. Meanwhile, MDSCs could make close interaction with osteoclasts, osteoblasts, chondrocytes, and other stromal cells within microenvironment of bone and joint, and thereby contributing to poor prognosis of bone-related diseases such as cancer-related bone metastasis, osteosarcoma (OS), rheumatoid arthritis (RA), osteoarthritis (OA), and orthopedic trauma. In addition, MDSCs have been shown to participate in the procedure of bone repair. In this review, we have summarized the function of MDSCs in cancer-related bone metastasis, the interaction with stromal cells within the bone microenvironment as well as joint microenvironment, and the critical role of MDSCs in bone repair. Besides, the promising value of MDSCs in the treatment for bone-related diseases is also well discussed.


Subject(s)
Bone Neoplasms/pathology , Bone Regeneration/physiology , Immune Tolerance/immunology , Myeloid-Derived Suppressor Cells/cytology , Arthritis, Rheumatoid/pathology , Bone Diseases/pathology , Bone Neoplasms/secondary , Cellular Microenvironment/physiology , Dendritic Cells/cytology , Granulocytes/cytology , Humans , Immunity, Innate/immunology , Macrophages/cytology , Myeloid-Derived Suppressor Cells/immunology , Osteoarthritis/pathology
16.
Lipids Health Dis ; 21(1): 5, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34996476

ABSTRACT

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.


Subject(s)
Bone and Bones/metabolism , Lipid Metabolism , Animals , Bone Diseases/metabolism , Bone Diseases/physiopathology , Bone Neoplasms/metabolism , Bone Neoplasms/physiopathology , Bone and Bones/physiology , Bone and Bones/physiopathology , Cellular Microenvironment/physiology , Cholesterol/metabolism , Cholesterol/physiology , Humans , Lipid Metabolism/physiology , Osteoporosis/metabolism
17.
Proc Natl Acad Sci U S A ; 116(51): 25555-25561, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31776251

ABSTRACT

Human population growth, soil degradation, and agrochemical misuse are significant challenges that agriculture must face in the upcoming decades as it pertains to global food production. Seed enhancement technologies will play a pivotal role in supporting food security by enabling germination of seeds in degraded environments, reducing seed germination time, and boosting crop yields. So far, a great effort has been pursued in designing plants that can adapt to different environments and germinate in the presence of abiotic stressors, such as soil salinity, heat, and drought. The technology proposed here seeks a different goal: To engineer the microenvironment of seeds by encapsulation, preservation, and precise delivery of biofertilizers that can boost seed germination and mitigate abiotic stressors. In particular, we developed a biomaterial based on silk fibroin (S) and trehalose that can be mixed with rhizobacteria and applied on the surface of seeds, retrofitting currently used techniques for seed coating, i.e., dip coating or spray drying. A micrometer thick transparent robust coating is formed by material assembly. The combination of a polymorphic protein as S and of a disaccharide used by living systems to tolerate abiotic stressors provides a beneficial environment for the survival of nonspore forming rhizobacteria outside the soil and in anhydrous conditions. Using Rhizobium tropici CIAT 899 and Phaseolus vulgaris as working models, we demonstrated that rhizobacteria delivered in the soil after coating dissolution infect seedlings' roots, form root nodules, enhance yield, boost germination, and mitigate soil salinity.


Subject(s)
Bioengineering/methods , Cellular Microenvironment/physiology , Germination/physiology , Seeds/physiology , Biocompatible Materials/chemistry , Fibroins/chemistry , Phaseolus , Plant Roots/physiology , Rhizobium , Salt Tolerance/physiology , Soil Microbiology , Trehalose/chemistry
18.
Int J Mol Sci ; 23(4)2022 Feb 11.
Article in English | MEDLINE | ID: mdl-35216117

ABSTRACT

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.


Subject(s)
Adipose Tissue/physiology , Cellular Microenvironment/physiology , Mesenchymal Stem Cells/physiology , Mitochondria/physiology , Adipocytes/physiology , Carcinogenesis/pathology , Cell Count/methods , Cell Line , Coculture Techniques/methods , HEK293 Cells , Humans
19.
J Mol Cell Cardiol ; 159: 105-119, 2021 10.
Article in English | MEDLINE | ID: mdl-34118218

ABSTRACT

Controlling fibrosis is an essential part of regenerating the post-ischemic heart. In the post-ischemic heart, fibroblasts differentiate to myofibroblasts that produce collagen-rich matrix to physically stabilize the infarct area. Infarct models in adult mice result in permanent scarring unlike newborn animals which fully regenerate. Decellularized extracellular matrix (dECM) hydrogels derived from early-aged hearts have been shown to be a transplantable therapy that preserves heart function and stimulates cardiomyocyte proliferation and vascularization. In this study, we investigate the anti-fibrotic effects of injectable dECM hydrogels in a cardiac explant model in the context of age-associated tissue compliance. Treatments with adult and fetal dECM hydrogels were tested for molecular effects on cardiac fibroblast activation and fibrosis. Altered sensitivity of fibroblasts to the mechanosignaling of the remodeling microenvironment was evaluated by manipulating the native extracellular matrix in explants and also with elastomeric substrates in the presence of dECM hydrogels. The injectable fetal dECM hydrogel treatment decreases fibroblast activation and contractility and lowers the stiffness-mediated increases in fibroblast activation observed in stiffened explants. The anti-fibrotic effect of dECM hydrogel is most observable at highest stiffness. Experiments with primary cells on elastomeric substrates with dECM treatment support this phenomenon. Transcriptome analysis indicated that dECM hydrogels affect cytoskeleton related signaling including Macrophage capping protein (CAPG) and Leupaxin (LPXN). CAPG was down-regulated by the fetal dECM hydrogel. LPXN expression was decreased by stiffening the explants; however, this effect was reversed by dECM hydrogel treatment. Pharmacological disruption of cytoskeleton polymerization lowered fibroblast activation and CAPG levels. Knocking down CAPG expression with siRNA inhibited fibroblast activation and collagen deposition. Collectively, fibroblast activation is dependent on cooperative action of extracellular molecular signals and mechanosignaling by cytoskeletal integrity.


Subject(s)
Cellular Microenvironment/physiology , Decellularized Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Microfilament Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Collagen/metabolism , Fibrosis/metabolism , Heart/physiology , Mice , Regeneration/physiology
20.
J Cell Physiol ; 236(2): 741-762, 2021 02.
Article in English | MEDLINE | ID: mdl-32657458

ABSTRACT

In the last four decades, several researchers worldwide have routinely and meticulously exercised cell culture experiments in two-dimensional (2D) platforms. Using traditionally existing 2D models, the therapeutic efficacy of drugs has been inappropriately validated due to the failure in generating the precise therapeutic response. Fortunately, a 3D model addresses the foregoing limitations by recapitulating the in vivo environment. In this context, one has to contemplate the design of an appropriate scaffold for favoring the organization of cell microenvironment. Instituting pertinent model on the platter will pave way for a precise mimicking of in vivo conditions. It is because animal cells in scaffolds oblige spontaneous formation of 3D colonies that molecularly, phenotypically, and histologically resemble the native environment. The 3D culture provides insight into the biochemical aspects of cell-cell communication, plasticity, cell division, cytoskeletal reorganization, signaling mechanisms, differentiation, and cell death. Focusing on these criteria, this paper discusses in detail, the diversification of polymeric scaffolds based on their available resources. The paper also reviews the well-founded and latest techniques of scaffold fabrication, and their applications pertaining to tissue engineering, drug screening, and tumor model development.


Subject(s)
Biomimetics/methods , Cell Culture Techniques/methods , Tissue Scaffolds/chemistry , Animals , Cellular Microenvironment/physiology , Humans , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL