Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Adv Sci (Weinh) ; 11(32): e2400719, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39189477

ABSTRACT

Collective cell migration is a model for nonequilibrium biological dynamics, which is important for morphogenesis, pattern formation, and cancer metastasis. The current understanding of cellular collective dynamics is based primarily on cells moving within a 2D epithelial monolayer. However, solid tumors often invade surrounding tissues in the form of a stream-like 3D structure, and how biophysical cues are integrated at the cellular level to give rise to this collective streaming remains unclear. Here, it is shown that cell cycle-mediated bioenergetics drive a forward advective flow of cells and energy to the front to support 3D collective invasion. The cell division cycle mediates a corresponding energy cycle such that cellular adenosine triphosphate (ATP) energy peaks just before division. A reaction-advection-diffusion (RAD) type model coupled with experimental measurements further indicates that most cells enter an active division cycle at rear positions during 3D streaming. Once the cells progress to a later stage toward division, the high intracellular energy allows them to preferentially stream toward the tip and become leader cells. This energy-driven cellular flow may be a fundamental characteristic of 3D collective dynamics based on thermodynamic principles important for not only cancer invasion but also tissue morphogenesis.


Subject(s)
Cell Movement , Energy Metabolism , Humans , Cell Movement/physiology , Energy Metabolism/physiology , Neoplasm Invasiveness , Cell Cycle/physiology , Adenosine Triphosphate/metabolism , Models, Biological , Cell Line, Tumor
2.
Matrix Biol ; 133: 77-85, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39147247

ABSTRACT

To form blood vessels, endothelial cells rearrange their cytoskeleton, generate traction stresses, migrate, and proliferate, all of which require energy. Despite these energetic costs, stiffening of the extracellular matrix promotes tumor angiogenesis and increases cell contractility. However, the interplay between extracellular matrix, cell contractility, and cellular energetics remains mechanistically unclear. Here, we utilized polyacrylamide substrates with various stiffnesses, a real-time biosensor of ATP, and traction force microscopy to show that endothelial cells exhibit increasing traction forces and energy usage trend as substrate stiffness increases. Inhibition of cytoskeleton reorganization via ROCK inhibition resulted in decreased cellular energy efficiency, and an opposite trend was found when cells were treated with manganese to promote integrin affinity. Altogether, our data reveal a link between matrix stiffness, cell contractility, and cell energetics, suggesting that endothelial cells on stiffer substrates can better convert intracellular energy into cellular traction forces. Given the critical role of cellular metabolism in cell function, our study also suggests that not only energy production but also the efficiency of its use plays a vital role in regulating cell behaviors and may help explain how increased matrix stiffness promotes angiogenesis.


Subject(s)
Acrylic Resins , Cytoskeleton , Endothelial Cells , Extracellular Matrix , Extracellular Matrix/metabolism , Humans , Acrylic Resins/chemistry , Endothelial Cells/metabolism , Cytoskeleton/metabolism , Energy Metabolism , Adenosine Triphosphate/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , rho-Associated Kinases/metabolism , rho-Associated Kinases/genetics , rho-Associated Kinases/antagonists & inhibitors , Integrins/metabolism , Integrins/genetics , Manganese/metabolism , Cell Movement , Cell Adhesion
3.
Cell Rep ; 42(4): 112338, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37027295

ABSTRACT

During intravasation, cancer cells cross the endothelial barrier and enter the circulation. Extracellular matrix stiffening has been correlated with tumor metastatic potential; however, little is known about the effects of matrix stiffness on intravasation. Here, we utilize in vitro systems, a mouse model, specimens from patients with breast cancer, and RNA expression profiles from The Cancer Genome Atlas Program (TCGA) to investigate the molecular mechanism by which matrix stiffening promotes tumor cell intravasation. Our data show that heightened matrix stiffness increases MENA expression, which promotes contractility and intravasation through focal adhesion kinase activity. Further, matrix stiffening decreases epithelial splicing regulatory protein 1 (ESRP1) expression, which triggers alternative splicing of MENA, decreases the expression of MENA11a, and enhances contractility and intravasation. Altogether, our data indicate that matrix stiffness regulates tumor cell intravasation through enhanced expression and ESRP1-mediated alternative splicing of MENA, providing a mechanism by which matrix stiffness regulates tumor cell intravasation.


Subject(s)
Alternative Splicing , Breast Neoplasms , Animals , Female , Humans , Mice , Alternative Splicing/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , RNA-Binding Proteins/metabolism
4.
Acta Biomater ; 163: 365-377, 2023 06.
Article in English | MEDLINE | ID: mdl-35483629

ABSTRACT

The role of intratumor heterogeneity is becoming increasingly apparent in part due to expansion in single cell technologies. Clinically, tumor heterogeneity poses several obstacles to effective cancer therapy dealing with biomarker variability and treatment responses. Matrix stiffening is known to occur during tumor progression and contribute to pathogenesis in several cancer hallmarks, including tumor angiogenesis and metastasis. However, the effects of matrix stiffening on intratumor heterogeneity have not been thoroughly studied. In this study, we applied single-cell RNA sequencing to investigate the differences in the transcriptional landscapes between stiff and compliant MMTV-PyMT mouse mammary tumors. We found similar compositions of cancer and stromal subpopulations in compliant and stiff tumors but differential intercellular communication and a significantly higher concentration of tumor-promoting, M2-like macrophages in the stiffer tumor microenvironments. Interestingly, we found that cancer cells seeded on stiffer substrates recruited more macrophages. Furthermore, elevated matrix stiffness increased Colony Stimulating Factor 1 (CSF-1) expression in breast cancer cells and reduction of CSF-1 expression on stiffer substrates reduced macrophage recruitment. Thus, our results demonstrate that tissue phenotypes were conserved between stiff and compliant tumors but matrix stiffening altered cell-cell interactions which may be responsible for shifting the phenotypic balance of macrophages residing in the tumor microenvironment towards a pro-tumor progression M2 phenotype. STATEMENT OF SIGNIFICANCE: Cells within tumors are highly heterogeneous, posing challenges with treatment and recurrence. While increased tissue stiffness can promote several hallmarks of cancer, its effects on tumor heterogeneity are unclear. We used single-cell RNA sequencing to investigate the differences in the transcriptional landscapes between stiff and compliant MMTV-PyMT mouse mammary tumors. We found similar compositions of cancer and stromal subpopulations in compliant and stiff tumors but differential intercellular communication and a significantly higher concentration of tumor-promoting, M2-like macrophages in the stiffer tumor microenvironments. Using a biomaterial-based platform, we found that cancer cells seeded on stiffer substrates recruited more macrophages, supporting our in vivo findings. Together, our results demonstrate a key role of matrix stiffness in affecting cell-cell communication and macrophage recruitment.


Subject(s)
Macrophage Colony-Stimulating Factor , Mammary Neoplasms, Animal , Animals , Mice , Macrophage Colony-Stimulating Factor/metabolism , Tumor Microenvironment , Macrophages/metabolism , Cell Communication , Mammary Neoplasms, Animal/pathology , Cell Line, Tumor
5.
Sci Adv ; 8(46): eabo1673, 2022 11 16.
Article in English | MEDLINE | ID: mdl-36399580

ABSTRACT

Diabetes mellitus is a complex metabolic disorder that is associated with an increased risk of breast cancer. Despite this correlation, the interplay between tumor progression and diabetes, particularly with regard to stiffening of the extracellular matrix, is still mechanistically unclear. Here, we established a murine model where hyperglycemia was induced before breast tumor development. Using the murine model, in vitro systems, and patient samples, we show that hyperglycemia increases tumor growth, extracellular matrix stiffness, glycation, and epithelial-mesenchymal transition of tumor cells. Upon inhibition of glycation or mechanotransduction in diabetic mice, these same metrics are reduced to levels comparable with nondiabetic tumors. Together, our study describes a novel biomechanical mechanism by which diabetic hyperglycemia promotes breast tumor progression via glycating the extracellular matrix. In addition, our work provides evidence that glycation inhibition is a potential adjuvant therapy for diabetic cancer patients due to the key role of matrix stiffening in both diseases.


Subject(s)
Diabetes Mellitus, Experimental , Hyperglycemia , Neoplasms , Mice , Animals , Mechanotransduction, Cellular , Disease Models, Animal , Extracellular Matrix/metabolism , Neoplasms/metabolism
6.
Soft Matter ; 18(44): 8504-8513, 2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36325938

ABSTRACT

Diabetes is associated with increased risk of breast cancer and worse prognoses for cancer patients. Hyperglycemia can result in increased glycation, the process wherein crosslinkages are formed between sugars and extracellular matrix (ECM) proteins through the formation of advanced glycation endproducts (AGEs). Although accumulation of AGEs occurs naturally in vivo over time, it is greatly accelerated by the hyperglycemic environment of diabetic patients. AGE accumulation has been linked to stiffening-related diseases such as hypertension, cancer metastasis, and neurodegenerative disorders. In response, several AGE-inhibiting and AGE-breaking drugs have received significant attention for their ability to reduce AGE accumulation. The resulting effects of these drugs on cell behavior is not well understood. In this study, we measured cancer cell migration in glycated collagen with and without the AGE-breaking drug alagebrium chloride (ALT711) to investigate the drug's ability to disrupt ECM crosslinks and reduce tumor cell spreading, contractility, and migration. The mechanical properties and chemical composition of collagen glycated with increasing concentrations of glucose with and without ALT711 treatment were measured. Increasing glucose concentration resulted in increased AGE accumulation and matrix stiffness as well as increased cancer cell contractility, elongation, and migration. Treatment with ALT711 significantly lowered AGE accumulation within the collagen, decreased collagen stiffness, and reduced cell migration. These findings suggest that while hyperglycemia can increase collagen matrix stiffness, resulting in increased breast cancer cell migration, an AGE-breaker can reverse this phenotype and may be a viable treatment option for reducing cancer cell migration due to glycation.


Subject(s)
Hyperglycemia , Neoplasms , Humans , Glycation End Products, Advanced/metabolism , Cell Movement , Collagen/metabolism , Glucose , Neoplasms/drug therapy
7.
iScience ; 25(10): 105190, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36274934

ABSTRACT

Intracellular and environmental cues result in heterogeneous cancer cell populations with different metabolic and migratory behaviors. Although glucose metabolism and epithelial-to-mesenchymal transition have previously been linked, we aim to understand how this relationship fuels cancer cell migration. We show that while glycolysis drives single-cell migration in confining microtracks, fast and slow cells display different migratory sensitivities to glycolysis and oxidative phosphorylation inhibition. Phenotypic sorting of highly and weakly migratory subpopulations (MDA+, MDA-) reveals that more mesenchymal, highly migratory MDA+ preferentially use glycolysis while more epithelial, weakly migratory MDA- utilize mitochondrial respiration. These phenotypes are plastic and MDA+ can be made less glycolytic, mesenchymal, and migratory and MDA- can be made more glycolytic, mesenchymal, and migratory via modulation of glucose metabolism or EMT. These findings reveal an intrinsic link between EMT and glucose metabolism that controls migration. Identifying mechanisms fueling phenotypic heterogeneity is essential to develop targeted metastatic therapeutics.

8.
Trends Cancer ; 7(10): 883-885, 2021 10.
Article in English | MEDLINE | ID: mdl-34417163

ABSTRACT

Focus on metabolic reprogramming has re-emerged in recent years due to the far-reaching consequences of metabolism on nearly all cellular behaviors. In a recent study in Cell Metabolism, Tharp et al. show that adhesion-dependent mechanical signaling induces mitochondrial and metabolic reprogramming to help cells adapt to future oxidative stress.


Subject(s)
Mitochondria , Oxidative Stress , Mechanotransduction, Cellular , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
9.
Cancer Res ; 81(13): 3649-3663, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33975882

ABSTRACT

Although intratumoral genomic heterogeneity can impede cancer research and treatment, less is known about the effects of phenotypic heterogeneities. To investigate the role of cell migration heterogeneities in metastasis, we phenotypically sorted metastatic breast cancer cells into two subpopulations based on migration ability. Although migration is typically considered to be associated with metastasis, when injected orthotopically in vivo, the weakly migratory subpopulation metastasized significantly more than the highly migratory subpopulation. To investigate the mechanism behind this observation, both subpopulations were assessed at each stage of the metastatic cascade, including dissemination from the primary tumor, survival in the circulation, extravasation, and colonization. Although both subpopulations performed each step successfully, weakly migratory cells presented as circulating tumor cell (CTC) clusters in the circulation, suggesting clustering as one potential mechanism behind the increased metastasis of weakly migratory cells. RNA sequencing revealed weakly migratory subpopulations to be more epithelial and highly migratory subpopulations to be more mesenchymal. Depletion of E-cadherin expression from weakly migratory cells abrogated metastasis. Conversely, induction of E-cadherin expression in highly migratory cells increased metastasis. Clinical patient data and blood samples showed that CTC clustering and E-cadherin expression are both associated with worsened patient outcome. This study demonstrates that deconvolving phenotypic heterogeneities can reveal fundamental insights into metastatic progression. More specifically, these results indicate that migratory ability does not necessarily correlate with metastatic potential and that E-cadherin promotes metastasis in phenotypically sorted breast cancer cell subpopulations by enabling CTC clustering. SIGNIFICANCE: This study employs phenotypic cell sorting for migration to reveal a weakly migratory, highly metastatic breast cancer cell subpopulation regulated by E-cadherin, highlighting the dichotomy between cancer cell migration and metastasis.


Subject(s)
Antigens, CD/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Neoplastic Cells, Circulating/pathology , Animals , Antigens, CD/genetics , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cadherins/genetics , Cell Proliferation , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Neoplastic Cells, Circulating/metabolism , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Nat Metab ; 2(11): 1248-1264, 2020 11.
Article in English | MEDLINE | ID: mdl-33106689

ABSTRACT

In addition to fatty acids, glucose and lactate are important myocardial substrates under physiologic and stress conditions. They are metabolized to pyruvate, which enters mitochondria via the mitochondrial pyruvate carrier (MPC) for citric acid cycle metabolism. In the present study, we show that MPC-mediated mitochondrial pyruvate utilization is essential for the partitioning of glucose-derived cytosolic metabolic intermediates, which modulate myocardial stress adaptation. Mice with cardiomyocyte-restricted deletion of subunit 1 of MPC (cMPC1-/-) developed age-dependent pathologic cardiac hypertrophy, transitioning to a dilated cardiomyopathy and premature death. Hypertrophied hearts accumulated lactate, pyruvate and glycogen, and displayed increased protein O-linked N-acetylglucosamine, which was prevented by increasing availability of non-glucose substrates in vivo by a ketogenic diet (KD) or a high-fat diet, which reversed the structural, metabolic and functional remodelling of non-stressed cMPC1-/- hearts. Although concurrent short-term KDs did not rescue cMPC1-/- hearts from rapid decompensation and early mortality after pressure overload, 3 weeks of a KD before transverse aortic constriction was sufficient to rescue this phenotype. Together, our results highlight the centrality of pyruvate metabolism to myocardial metabolism and function.


Subject(s)
Adaptation, Physiological/physiology , Anion Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Myocardium/metabolism , Stress, Physiological/physiology , Adaptation, Physiological/genetics , Animals , Anion Transport Proteins/genetics , Cardiomegaly/diagnostic imaging , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Constriction, Pathologic , Cytosol/metabolism , Diet, High-Fat , Diet, Ketogenic , Echocardiography , In Vitro Techniques , Mice , Mice, Knockout , Mitochondria, Heart/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Myocytes, Cardiac/metabolism , Pyruvic Acid/metabolism , Stress, Physiological/genetics
12.
Nat Commun ; 10(1): 5619, 2019 Dec 04.
Article in English | MEDLINE | ID: mdl-31797873

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

13.
Cell Mol Bioeng ; 12(1): 1-14, 2019 Feb.
Article in English | MEDLINE | ID: mdl-31565083

ABSTRACT

Communication between cancer cells enables cancer progression and metastasis. While cell-cell communication in cancer has primarily been examined through chemical mechanisms, recent evidence suggests that mechanical communication through cell-cell junctions and cell-ECM linkages is also an important mediator of cancer progression. Cancer and stromal cells remodel the ECM through a variety of mechanisms, including matrix degradation, cross-linking, deposition, and physical remodeling. Cancer cells sense these mechanical environmental changes through cell-matrix adhesion complexes and subsequently alter their tension between both neighboring cells and the surrounding matrix, thereby altering the force landscape within the microenvironment. This communication not only allows cancer cells to communicate with each other, but allows stromal cells to communicate with cancer cells through matrix remodeling. Here, we review the mechanisms of intercellular force transmission, the subsequent matrix remodeling, and the implications of this mechanical communication on cancer progression.

14.
Nat Commun ; 10(1): 4185, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31519914

ABSTRACT

Cell migration during the invasion-metastasis cascade requires cancer cells to navigate a spatially complex microenvironment that presents directional choices to migrating cells. Here, we investigate cellular energetics during migration decision-making in confined spaces. Theoretical and experimental data show that energetic costs for migration through confined spaces are mediated by a balance between cell and matrix compliance as well as the degree of spatial confinement to direct decision-making. Energetic costs, driven by the cellular work needed to generate force for matrix displacement, increase with increasing cell stiffness, matrix stiffness, and degree of spatial confinement, limiting migration. By assessing energetic costs between possible migration paths, we can predict the probability of migration choice. Our findings indicate that motility in confined spaces imposes high energetic demands on migrating cells, and cells migrate in the direction of least confinement to minimize energetic costs. Therefore, therapeutically targeting metabolism may limit cancer cell migration and metastasis.


Subject(s)
Cell Movement/physiology , Decision Making , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Biomedical Engineering , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line, Tumor , Cell Movement/genetics , Glucose/metabolism , Humans , Microscopy, Atomic Force , Microscopy, Confocal , Microscopy, Phase-Contrast , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
15.
PLoS One ; 14(5): e0216537, 2019.
Article in English | MEDLINE | ID: mdl-31091287

ABSTRACT

Aligned collagen architecture is a characteristic feature of the tumor extracellular matrix (ECM) and has been shown to facilitate cancer metastasis using 3D in vitro models. Additional features of the ECM, such as pore size and stiffness, have also been shown to influence cellular behavior and are implicated in cancer progression. While there are several methods to produce aligned matrices to study the effect on cell behavior in vitro, it is unclear how the alignment itself may alter these other important features of the matrix. In this study, we have generated aligned collagen matrices and characterized their pore sizes and mechanical properties at the micro- and macro-scale. Our results indicate that collagen alignment can alter pore-size of matrices depending on the polymerization temperature of the collagen. Furthermore, alignment does not affect the macro-scale stiffness but alters the micro-scale stiffness in a temperature independent manner. Overall, these results describe the manifestation of confounding variables that arise due to alignment and the importance of fully characterizing biomaterials at both micro- and macro-scales.


Subject(s)
Extracellular Matrix/pathology , Fibrillar Collagens/metabolism , Neoplasms/diagnostic imaging , Algorithms , Cell Movement , Disease Progression , Extracellular Matrix/metabolism , Humans , Microscopy, Atomic Force , Neoplasms/metabolism
16.
Proc Natl Acad Sci U S A ; 116(16): 7867-7872, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30923113

ABSTRACT

The ability of primary tumor cells to invade into adjacent tissues, followed by the formation of local or distant metastasis, is a lethal hallmark of cancer. Recently, locomoting clusters of tumor cells have been identified in numerous cancers and associated with increased invasiveness and metastatic potential. However, how the collective behaviors of cancer cells are coordinated and their contribution to cancer invasion remain unclear. Here we show that collective invasion of breast cancer cells is regulated by the energetic statuses of leader and follower cells. Using a combination of in vitro spheroid and ex vivo organoid invasion models, we found that cancer cells dynamically rearrange leader and follower positions during collective invasion. Cancer cells invade cooperatively in denser collagen matrices by accelerating leader-follower switching thus decreasing leader cell lifetime. Leader cells exhibit higher glucose uptake than follower cells. Moreover, their energy levels, as revealed by the intracellular ATP/ADP ratio, must exceed a threshold to invade. Forward invasion of the leader cell gradually depletes its available energy, eventually leading to leader-follower transition. Our computational model based on intracellular energy homeostasis successfully recapitulated the dependence of leader cell lifetime on collagen density. Experiments further supported model predictions that decreasing the cellular energy level by glucose starvation decreases leader cell lifetime whereas increasing the cellular energy level by AMP-activated kinase (AMPK) activation does the opposite. These findings highlight coordinated invasion and its metabolic regulation as potential therapeutic targets of cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Cell Movement/physiology , Energy Metabolism/physiology , Neoplasm Invasiveness/physiopathology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Female , Glucose/metabolism , Humans , Intracellular Space/metabolism
17.
Integr Biol (Camb) ; 10(12): 734-746, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30382278

ABSTRACT

Intimal stiffening has been linked with increased vascular permeability and leukocyte transmigration, hallmarks of atherosclerosis. However, recent evidence indicates age-related intimal stiffening is not uniform but rather characterized by increased point-to-point heterogeneity in subendothelial matrix stiffness, the impact of which is much less understood. To investigate the impact of spatially heterogeneous matrix rigidity on endothelial monolayer integrity, we develop a micropillar model to introduce closely-spaced, step-changes in substrate rigidity and compare endothelial monolayer phenotype to rigidity-matched, uniformly stiff and compliant substrates. We found equivalent disruption of adherens junctions within monolayers on step-rigidity and uniformly stiff substrates relative to uniformly compliant substrates. Similarly, monolayers cultured on step-rigidity substrates exhibited equivalent percentages of leukocyte transmigration to monolayers on rigidity-matched, uniformly stiff substrates. Adherens junction tension and focal adhesion density, but not size, increased within monolayers on step-rigidity and uniformly stiff substrates compared to more compliant substrates suggesting that elevated tension is disrupting adherens junction integrity. Leukocyte transmigration frequency and time, focal adhesion size, and focal adhesion density did not differ between stiff and compliant sub-regions of step-rigidity substrates. Overall, our results suggest that endothelial monolayers exposed to mechanically heterogeneous substrates adopt the phenotype associated with the stiffer matrix, indicating that spatial heterogeneities in intimal stiffness observed with age could disrupt endothelial barrier integrity and contribute to atherogenesis.


Subject(s)
Atherosclerosis/physiopathology , Tunica Intima/pathology , Vascular Stiffness , Adherens Junctions , Animals , Aorta/pathology , Cattle , Cell Adhesion , Cell Communication , Cell Movement , Dimethylpolysiloxanes/chemistry , Endothelial Cells/cytology , Endothelium, Vascular/pathology , Focal Adhesions/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Leukocytes/cytology , Materials Testing , Neutrophils/cytology , Phenotype , Vinculin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL