Catalogue Search | MBRL
Search Results Heading
Explore the vast range of titles available.
MBRLSearchResults
-
DisciplineDiscipline
-
Is Peer ReviewedIs Peer Reviewed
-
Item TypeItem Type
-
SubjectSubject
-
YearFrom:-To:
-
More FiltersMore FiltersSourceLanguage
Done
Filters
Reset
375
result(s) for
"Hepatic Stellate Cells - cytology"
Sort by:
Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations
2018
The liver is the largest solid organ in the body and is critical for metabolic and immune functions. However, little is known about the cells that make up the human liver and its immune microenvironment. Here we report a map of the cellular landscape of the human liver using single-cell RNA sequencing. We provide the transcriptional profiles of 8444 parenchymal and non-parenchymal cells obtained from the fractionation of fresh hepatic tissue from five human livers. Using gene expression patterns, flow cytometry, and immunohistochemical examinations, we identify 20 discrete cell populations of hepatocytes, endothelial cells, cholangiocytes, hepatic stellate cells, B cells, conventional and non-conventional T cells, NK-like cells, and distinct intrahepatic monocyte/macrophage populations. Together, our study presents a comprehensive view of the human liver at single-cell resolution that outlines the characteristics of resident cells in the liver, and in particular provides a map of the human hepatic immune microenvironment.
The development of single cell RNA sequencing technologies has been instrumental in advancing our understanding of tissue biology. Here, MacParland et al. performed single cell RNA sequencing of human liver samples, and identify distinct populations of intrahepatic macrophages that may play specific roles in liver disease.
Journal Article
Resolving the fibrotic niche of human liver cirrhosis at single-cell level
2019
Liver cirrhosis is a major cause of death worldwide and is characterized by extensive fibrosis. There are currently no effective antifibrotic therapies available. To obtain a better understanding of the cellular and molecular mechanisms involved in disease pathogenesis and enable the discovery of therapeutic targets, here we profile the transcriptomes of more than 100,000 single human cells, yielding molecular definitions for non-parenchymal cell types that are found in healthy and cirrhotic human liver. We identify a scar-associated TREM2
+
CD9
+
subpopulation of macrophages, which expands in liver fibrosis, differentiates from circulating monocytes and is pro-fibrogenic. We also define ACKR1
+
and PLVAP
+
endothelial cells that expand in cirrhosis, are topographically restricted to the fibrotic niche and enhance the transmigration of leucocytes. Multi-lineage modelling of ligand and receptor interactions between the scar-associated macrophages, endothelial cells and PDGFRα
+
collagen-producing mesenchymal cells reveals intra-scar activity of several pro-fibrogenic pathways including TNFRSF12A, PDGFR and NOTCH signalling. Our work dissects unanticipated aspects of the cellular and molecular basis of human organ fibrosis at a single-cell level, and provides a conceptual framework for the discovery of rational therapeutic targets in liver cirrhosis.
Single-cell RNA sequencing is used to characterize and compare the functional diversity of cells from liver biopsies of human scarred and normal liver, and identifies markers for scar-associated macrophages and endothelial cells.
Journal Article
Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome
by
Ishikawa, Yuichi
,
Atarashi, Koji
,
Sato, Seidai
in
631/67/70
,
Animals
,
Anti-Bacterial Agents - pharmacology
2013
Obesity is shown in a mouse model of liver cancer to strongly enhance tumorigenesis; a high fat diet alters the composition of intestinal bacteria, leading to more production of the metabolite DCA which, probably together with other factors, induces senescence and the secretion of various senescence-associated cytokines in hepatic stellate cells, thus promoting cancer.
Bile acid metabolite links diet and cancer
Epidemiological data have demonstrated a link between obesity and cancer. This study shows that in a mouse model of liver cancer, a high-fat diet strongly enhances tumorigenesis by provoking a senescence-associated secretory phenotype (SASP), a recently identified senescent phenotype associated with the secretion of various tumour-promoting factors. Antibiotic and other interventions show that the fatty diet altered the composition of intestinal bacteria leading to more production of deoxycholic acid (DCA), a by-product of microbial bile acid metabolism that is known to cause DNA damage. The authors suggest that DCA, acting with other as-yet unknown factors, induces senescence and the secretion of various senescence-associated cytokines in hepatic stellate cells. These cytokines in turn act to promote the development of liver cancer. These findings highlight the complex mechanistic links between diet, the microbiota and cancer and suggest novel therapeutic approaches.
Obesity has become more prevalent in most developed countries over the past few decades, and is increasingly recognized as a major risk factor for several common types of cancer
1
. As the worldwide obesity epidemic has shown no signs of abating
2
, better understanding of the mechanisms underlying obesity-associated cancer is urgently needed. Although several events were proposed to be involved in obesity-associated cancer
1
,
3
, the exact molecular mechanisms that integrate these events have remained largely unclear. Here we show that senescence-associated secretory phenotype (SASP)
4
,
5
has crucial roles in promoting obesity-associated hepatocellular carcinoma (HCC) development in mice. Dietary or genetic obesity induces alterations of gut microbiota, thereby increasing the levels of deoxycholic acid (DCA), a gut bacterial metabolite known to cause DNA damage
6
. The enterohepatic circulation of DCA provokes SASP phenotype in hepatic stellate cells (HSCs)
7
, which in turn secretes various inflammatory and tumour-promoting factors in the liver, thus facilitating HCC development in mice after exposure to chemical carcinogen. Notably, blocking DCA production or reducing gut bacteria efficiently prevents HCC development in obese mice. Similar results were also observed in mice lacking an SASP inducer
8
or depleted of senescent HSCs, indicating that the DCA–SASP axis in HSCs has key roles in obesity-associated HCC development. Moreover, signs of SASP were also observed in the HSCs in the area of HCC arising in patients with non-alcoholic steatohepatitis
3
, indicating that a similar pathway may contribute to at least certain aspects of obesity-associated HCC development in humans as well. These findings provide valuable new insights into the development of obesity-associated cancer and open up new possibilities for its control.
Journal Article
The Role of Fibrosis and Liver-Associated Fibroblasts in the Pathogenesis of Hepatocellular Carcinoma
2019
Hepatocellular carcinoma (HCC) is one of the most aggressive types of cancer and lacks effective therapeutic approaches. Most HCC develops in the setting of chronic liver injury, hepatic inflammation, and fibrosis. Hepatic stellate cells (HSCs) and cancer-associated fibroblasts (CAFs) are key players in liver fibrogenesis and hepatocarcinogenesis, respectively. CAFs, which probably derive from HSCs, activate into extracellular matrix (ECM)-producing myofibroblasts and crosstalk with cancer cells to affect tumor growth and invasion. In this review, we describe the different components which form the HCC premalignant microenvironment (PME) and the tumor microenvironment (TME), focusing on the liver fibrosis process and the biology of CAFs. We will describe the CAF-dependent mechanisms which have been suggested to promote hepatocarcinogenesis, such as the alteration of ECM, CAF-dependent production of cytokines and angiogenic factors, CAF-dependent reduction of immuno-surveillance, and CAF-dependent promotion of epithelial-mesenchymal transition (EMT). New knowledge of the fibrosis process and the role of CAFs in HCC may pave the way for new therapeutic strategies for liver cancer.
Journal Article
Hepatic stellate cells suppress NK cell-sustained breast cancer dormancy
2021
The persistence of undetectable disseminated tumour cells (DTCs) after primary tumour resection poses a major challenge to effective cancer treatment
1
–
3
. These enduring dormant DTCs are seeds of future metastases, and the mechanisms that switch them from dormancy to outgrowth require definition. Because cancer dormancy provides a unique therapeutic window for preventing metastatic disease, a comprehensive understanding of the distribution, composition and dynamics of reservoirs of dormant DTCs is imperative. Here we show that different tissue-specific microenvironments restrain or allow the progression of breast cancer in the liver—a frequent site of metastasis
4
that is often associated with a poor prognosis
5
. Using mouse models, we show that there is a selective increase in natural killer (NK) cells in the dormant milieu. Adjuvant interleukin-15-based immunotherapy ensures an abundant pool of NK cells that sustains dormancy through interferon-γ signalling, thereby preventing hepatic metastases and prolonging survival. Exit from dormancy follows a marked contraction of the NK cell compartment and the concurrent accumulation of activated hepatic stellate cells (aHSCs). Our proteomics studies on liver co-cultures implicate the aHSC-secreted chemokine CXCL12 in the induction of NK cell quiescence through its cognate receptor CXCR4. CXCL12 expression and aHSC abundance are closely correlated in patients with liver metastases. Our data identify the interplay between NK cells and aHSCs as a master switch of cancer dormancy, and suggest that therapies aimed at normalizing the NK cell pool might succeed in preventing metastatic outgrowth.
Liver-resident natural killer (NK) cells sustain the dormancy of disseminated breast cancer cells, and a decrease in NK cells and increase in activated hepatic stellate cells is associated with the formation of liver metastases.
Journal Article
Liver fibrosis and repair: immune regulation of wound healing in a solid organ
by
Pellicoro, Antonella
,
Iredale, John P.
,
Fallowfield, Jonathan A.
in
631/250/2504/342
,
631/250/256/2515
,
692/699/1503/1607/1605
2014
Key Points
The liver provides a useful generic model of inflammation and repair, showing the complex interplay between the epithelial, inflammatory, myofibroblast and extracellular matrix (ECM) components of the mammalian wound-healing response. In almost all situations, fibrosis is preceded by inflammation and elements of both the innate and adaptive immune systems are crucial in regulating the fibrotic process.
Following liver injury, pro-inflammatory mediators that are generated by cellular damage and stimulated immune cells, as well as growth factors and cytokines (including platelet-derived growth factor, connective tissue growth factor, transforming growth factor-β and interleukin-13), activate mesenchymal precursor cells in tissues and induce their transdifferentiation into myofibroblasts. Myofibroblasts are master regulators of the fibrotic response as a result of their acquisition of scar-producing, proliferative, migratory, contractile, immunomodulatory and phagocytic properties.
Recent studies have used bone marrow transplantation techniques in reporter mice to show that, regardless of the aetiology or the duration of the injury, liver myofibroblasts are almost exclusively derived from the activation of resident mesenchymal cells. Perpetuation of myofibroblast fibrogenic activity is mediated through several positive feedback loops, involving the autocrine and paracrine effects of cytokines and growth factors, and cell–cell and cell–matrix interactions. Myofibroblasts themselves function as innate immune cells.
The balance of T helper 1 (T
H
1) cell-mediated and T
H
2 cell-mediated adaptive immune responses, the influence of unconventional T cell subsets and the equilibrium between different pro-inflammatory (that is, pro-fibrotic) and pro-resolution macrophage populations determine whether the outcome of tissue injury is homeostatic and self-limited or whether it results in pathogenic scarring.
Liver fibrosis in rodents and humans is a dynamic, bidirectional process that has an inherent capacity for recovery and remodelling. The loss of myofibroblasts from the hepatic scar and a crucial switch in macrophage phenotype to a pro-resolution cell type are important events in the regression of liver fibrosis that facilitate remodelling of the ECM.
A considerable number of tractable therapeutic targets have been identified in liver fibrosis, but clinical trials of anti-fibrotic therapies have so far been unsuccessful. Identification of the core pathways in fibrosis is likely to yield greater success in clinical translation.
The immune regulation of liver fibrosis (particularly the distinct and opposing roles of macrophage subsets) provides an informative model of the endogenous mechanisms that mediate the resolution of fibrosis and the restoration of tissue homeostasis.
Fibrosis is a highly conserved and co-ordinated protective response to tissue injury. The interaction of multiple pathways, molecules and systems determines whether fibrosis is self-limiting and homeostatic, or whether it is uncontrolled and excessive. Immune cells have been identified as key players in this fibrotic cascade, with the capacity to exert either injury-inducing or repair-promoting effects. A multi-organ approach was recently suggested to identify the core and regulatory pathways in fibrosis, with the aim of integrating the wealth of information emerging from basic fibrosis research. In this Review, we focus on recent advances in liver fibrosis research as a paradigm for wound healing in solid organs and the role of the immune system in regulating and balancing this response.
Journal Article
Human hepatic stellate cell isolation and characterization
2018
The hepatic stellate cells (HSCs) localize at the space of Disse in the liver and have multiple functions. They are identified as the major contributor to hepatic fibrosis. Significant understanding of HSCs has been achieved using rodent models and isolated murine HSCs; as well as investigating human liver tissues and human HSCs. There is growing interest and need of translating rodent study findings to human HSCs and human liver diseases. However, species-related differences impose challenges on the translational research. In this review, we focus on the current information on human HSCs isolation methods, human HSCs markers, and established human HSC cell lines.
Journal Article
Stem Cell Therapy in Heart Diseases – Cell Types, Mechanisms and Improvement Strategies
by
Lemcke, Heiko
,
Müller, Paula
,
David, Robert
in
Bone marrow
,
Cardiac arrhythmia
,
Cardiac regeneration
2018
A large number of clinical trials have shown stem cell therapy to be a promising therapeutic approach for the treatment of cardiovascular diseases. Since the first transplantation into human patients, several stem cell types have been applied in this field, including bone marrow derived stem cells, cardiac progenitors as well as embryonic stem cells and their derivatives. However, results obtained from clinical studies are inconsistent and stem cell-based improvement of heart performance and cardiac remodeling was found to be quite limited. In order to optimize stem cell efficiency, it is crucial to elucidate the underlying mechanisms mediating the beneficial effects of stem cell transplantation. Based on these mechanisms, researchers have developed different improvement strategies to boost the potency of stem cell repair and to generate the “next generation” of stem cell therapeutics. Moreover, since cardiovascular diseases are complex disorders including several disease patterns and pathologic mechanisms it may be difficult to provide a uniform therapeutic intervention for all subgroups of patients. Therefore, future strategies should aim at more personalized SC therapies in which individual disease parameters influence the selection of optimal cell type, dosage and delivery approach.
Journal Article
Single-cell and bulk transcriptomics of the liver reveals potential targets of NASH with fibrosis
2021
Fibrosis is characterized by the excessive production of collagen and other extracellular matrix (ECM) components and represents a leading cause of morbidity and mortality worldwide. Previous studies of nonalcoholic steatohepatitis (NASH) with fibrosis were largely restricted to bulk transcriptome profiles. Thus, our understanding of this disease is limited by an incomplete characterization of liver cell types in general and hepatic stellate cells (HSCs) in particular, given that activated HSCs are the major hepatic fibrogenic cell population. To help fill this gap, we profiled 17,810 non-parenchymal cells derived from six healthy human livers. In conjunction with public single-cell data of fibrotic/cirrhotic human livers, these profiles enable the identification of potential intercellular signaling axes (e.g., ITGAV–LAMC1, TNFRSF11B–VWF and NOTCH2–DLL4) and master regulators (e.g.,
RUNX1
and
CREB3L1
) responsible for the activation of HSCs during fibrogenesis. Bulk RNA-seq data of NASH patient livers and rodent models for liver fibrosis of diverse etiologies allowed us to evaluate the translatability of candidate therapeutic targets for NASH-related fibrosis. We identified 61 liver fibrosis-associated genes (e.g.,
AEBP1, PRRX1
and
LARP6
) that may serve as a repertoire of translatable drug target candidates. Consistent with the above regulon results, gene regulatory network analysis allowed the identification of
CREB3L1
as a master regulator of many of the 61 genes. Together, this study highlights potential cell–cell interactions and master regulators that underlie HSC activation and reveals genes that may represent prospective hallmark signatures for liver fibrosis.
Journal Article
Evolving therapies for liver fibrosis
2013
Fibrosis is an intrinsic response to chronic injury, maintaining organ integrity when extensive necrosis or apoptosis occurs. With protracted damage, fibrosis can progress toward excessive scarring and organ failure, as in liver cirrhosis. To date, antifibrotic treatment of fibrosis represents an unconquered area for drug development, with enormous potential but also high risks. Preclinical research has yielded numerous targets for antifibrotic agents, some of which have entered early-phase clinical studies, but progress has been hampered due to the relative lack of sensitive and specific biomarkers to measure fibrosis progression or reversal. Here we focus on antifibrotic approaches for liver that address specific cell types and functional units that orchestrate fibrotic wound healing responses and have a sound preclinical database or antifibrotic activity in early clinical trials. We also touch upon relevant clinical study endpoints, optimal study design, and developments in fibrosis imaging and biomarkers.
Journal Article