Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Raymond Liang is active.

Publication


Featured researches published by Raymond Liang.


Development | 2014

Stem cells and the impact of ROS signaling.

Carolina L. Bigarella; Raymond Liang; Saghi Ghaffari

An appropriate balance between self-renewal and differentiation is crucial for stem cell function during both early development and tissue homeostasis throughout life. Recent evidence from both pluripotent embryonic and adult stem cell studies suggests that this balance is partly regulated by reactive oxygen species (ROS), which, in synchrony with metabolism, mediate the cellular redox state. In this Primer, we summarize what ROS are and how they are generated in the cell, as well as their downstream molecular targets. We then review recent findings that provide molecular insights into how ROS signaling can influence stem cell homeostasis and lineage commitment, and discuss the implications of this for reprogramming and stem cell ageing. We conclude that ROS signaling is an emerging key regulator of multiple stem cell populations.


Stem cell reports | 2014

Aging-like Phenotype and Defective Lineage Specification in SIRT1-Deleted Hematopoietic Stem and Progenitor Cells

Pauline Rimmele; Carolina L. Bigarella; Raymond Liang; Brigitte Izac; Rebeca Dieguez-Gonzalez; Gaetan Barbet; Michael J. Donovan; Carlo Brugnara; Julie Magarian Blander; David A. Sinclair; Saghi Ghaffari

Summary Aging hematopoietic stem cells (HSCs) exhibit defective lineage specification that is thought to be central to increased incidence of myeloid malignancies and compromised immune competence in the elderly. Mechanisms underlying these age-related defects remain largely unknown. We show that the deacetylase Sirtuin (SIRT)1 is required for homeostatic HSC maintenance. Differentiation of young SIRT1-deleted HSCs is skewed toward myeloid lineage associated with a significant decline in the lymphoid compartment, anemia, and altered expression of associated genes. Combined with HSC accumulation of damaged DNA and expression patterns of age-linked molecules, these have striking overlaps with aged HSCs. We further show that SIRT1 controls HSC homeostasis via the longevity transcription factor FOXO3. These findings suggest that SIRT1 is essential for HSC homeostasis and lineage specification. They also indicate that SIRT1 might contribute to delaying HSC aging.


Antioxidants & Redox Signaling | 2014

Stem cells, redox signaling, and stem cell aging.

Raymond Liang; Saghi Ghaffari

SIGNIFICANCE Functional stem cell decline has been postulated to result in loss of maintenance of tissue homeostasis leading to organismal decline and diseases of aging. RECENT ADVANCES Recent findings implicate redox metabolism in the control of stem cell pool and stem cell aging. Although reactive oxygen species (ROS) are better known for their damaging properties to DNA, proteins and lipids, recent findings suggest that ROS may also be an integral physiological mediator of cellular signaling in primary cells. CRITICAL ISSUES Here we review recent published work on major signaling pathways and transcription factors that are regulated by ROS and mediate ROS regulation of stem cell fate. We will specifically focus on how alterations in this regulation may be implicated in disease and particularly in diseases of stem cell aging. In general, based on the work described here we propose a model in which ROS function as stem cell rheostat. FUTURE DIRECTIONS Future work in elucidating how ROS control stem cell cycling, apoptotic machinery, and lineage determination should shed light on mechanisms whereby ROS may control stem cell aging.


EMBO Reports | 2015

Mitochondrial metabolism in hematopoietic stem cells requires functional FOXO3

Pauline Rimmele; Raymond Liang; Carolina L. Bigarella; Fatih Kocabas; Jingjing Xie; Madhavika N. Serasinghe; Jerry E. Chipuk; Hesham A. Sadek; Cheng Cheng Zhang; Saghi Ghaffari

Hematopoietic stem cells (HSC) are primarily dormant but have the potential to become highly active on demand to reconstitute blood. This requires a swift metabolic switch from glycolysis to mitochondrial oxidative phosphorylation. Maintenance of low levels of reactive oxygen species (ROS), a by‐product of mitochondrial metabolism, is also necessary for sustaining HSC dormancy. Little is known about mechanisms that integrate energy metabolism with hematopoietic stem cell homeostasis. Here, we identify the transcription factor FOXO3 as a new regulator of metabolic adaptation of HSC. ROS are elevated in Foxo3−/− HSC that are defective in their activity. We show that Foxo3−/− HSC are impaired in mitochondrial metabolism independent of ROS levels. These defects are associated with altered expression of mitochondrial/metabolic genes in Foxo3−/− hematopoietic stem and progenitor cells (HSPC). We further show that defects of Foxo3−/− HSC long‐term repopulation activity are independent of ROS or mTOR signaling. Our results point to FOXO3 as a potential node that couples mitochondrial metabolism with HSC homeostasis. These findings have critical implications for mechanisms that promote malignant transformation and aging of blood stem and progenitor cells.


American Journal of Hematology | 2014

FOXO3‐mTOR metabolic cooperation in the regulation of erythroid cell maturation and homeostasis

Xin Zhang; Genís Campreciós; Pauline Rimmele; Raymond Liang; Safak Yalcin; Sathish Kumar Mungamuri; Jeffrey Barminko; Valentina d'Escamard; Margaret H. Baron; Carlo Brugnara; Dmitri Papatsenko; Stefano Rivella; Saghi Ghaffari

Ineffective erythropoiesis is observed in many erythroid disorders including β‐thalassemia and anemia of chronic disease in which increased production of erythroblasts that fail to mature exacerbate the underlying anemias. As loss of the transcription factor FOXO3 results in erythroblast abnormalities similar to the ones observed in ineffective erythropoiesis, we investigated the underlying mechanisms of the defective Foxo3−/− erythroblast cell cycle and maturation. Here we show that loss of Foxo3 results in overactivation of the JAK2/AKT/mTOR signaling pathway in primary bone marrow erythroblasts partly mediated by redox modulation. We further show that hyperactivation of mTOR signaling interferes with cell cycle progression in Foxo3 mutant erythroblasts. Importantly, inhibition of mTOR signaling, in vivo or in vitro enhances significantly Foxo3 mutant erythroid cell maturation. Similarly, in vivo inhibition of mTOR remarkably improves erythroid cell maturation and anemia in a model of β‐thalassemia. Finally we show that FOXO3 and mTOR are likely part of a larger metabolic network in erythroblasts as together they control the expression of an array of metabolic genes some of which are implicated in erythroid disorders. These combined findings indicate that a metabolism‐mediated regulatory network centered by FOXO3 and mTOR control the balanced production and maturation of erythroid cells. They also highlight physiological interactions between these proteins in regulating erythroblast energy. Our results indicate that alteration in the function of this network might be implicated in the pathogenesis of ineffective erythropoiesis. Am. J. Hematol. 89:954–963, 2014.


Nucleic Acids Research | 2015

Stable heteroplasmy at the single-cell level is facilitated by intercellular exchange of mtDNA

Anitha Jayaprakash; Erica K. Benson; Swapna Gone; Raymond Liang; Jaehee Shim; Luca Lambertini; Masoud M. Toloue; Michael Wigler; Stuart A. Aaronson; Ravi Sachidanandam

Eukaryotic cells carry two genomes, nuclear (nDNA) and mitochondrial (mtDNA), which are ostensibly decoupled in their replication, segregation and inheritance. It is increasingly appreciated that heteroplasmy, the occurrence of multiple mtDNA haplotypes in a cell, plays an important biological role, but its features are not well understood. Accurately determining the diversity of mtDNA has been difficult, due to the relatively small amount of mtDNA in each cell (<1% of the total DNA), the intercellular variability of mtDNA content and mtDNA pseudogenes (Numts) in nDNA. To understand the nature of heteroplasmy, we developed Mseek, a novel technique to purify and sequence mtDNA. Mseek yields high purity (>90%) mtDNA and its ability to detect rare variants is limited only by sequencing depth, providing unprecedented sensitivity and specificity. Using Mseek, we confirmed the ubiquity of heteroplasmy by analyzing mtDNA from a diverse set of cell lines and human samples. Applying Mseek to colonies derived from single cells, we find heteroplasmy is stably maintained in individual daughter cells over multiple cell divisions. We hypothesized that the stability of heteroplasmy could be facilitated by intercellular exchange of mtDNA. We explicitly demonstrate this exchange by co-culturing cell lines with distinct mtDNA haplotypes. Our results shed new light on the maintenance of heteroplasmy and provide a novel platform to investigate features of heteroplasmy in normal and diseased states.


PLOS Genetics | 2015

A Systems Approach Identifies Essential FOXO3 Functions at Key Steps of Terminal Erythropoiesis

Raymond Liang; Genís Campreciós; Yan Kou; Kathleen E. McGrath; Roberta B. Nowak; Seana C. Catherman; Carolina L. Bigarella; Pauline Rimmele; Xin Zhang; Merlin Nithya Gnanapragasam; James J. Bieker; Dmitri Papatsenko; Avi Ma’ayan; Emery H. Bresnick; Velia M. Fowler; James Palis; Saghi Ghaffari

Circulating red blood cells (RBCs) are essential for tissue oxygenation and homeostasis. Defective terminal erythropoiesis contributes to decreased generation of RBCs in many disorders. Specifically, ineffective nuclear expulsion (enucleation) during terminal maturation is an obstacle to therapeutic RBC production in vitro. To obtain mechanistic insights into terminal erythropoiesis we focused on FOXO3, a transcription factor implicated in erythroid disorders. Using an integrated computational and experimental systems biology approach, we show that FOXO3 is essential for the correct temporal gene expression during terminal erythropoiesis. We demonstrate that the FOXO3-dependent genetic network has critical physiological functions at key steps of terminal erythropoiesis including enucleation and mitochondrial clearance processes. FOXO3 loss deregulated transcription of genes implicated in cell polarity, nucleosome assembly and DNA packaging-related processes and compromised erythroid enucleation. Using high-resolution confocal microscopy and imaging flow cytometry we show that cell polarization is impaired leading to multilobulated Foxo3 -/- erythroblasts defective in nuclear expulsion. Ectopic FOXO3 expression rescued Foxo3 -/- erythroblast enucleation-related gene transcription, enucleation defects and terminal maturation. Remarkably, FOXO3 ectopic expression increased wild type erythroblast maturation and enucleation suggesting that enhancing FOXO3 activity may improve RBCs production. Altogether these studies uncover FOXO3 as a novel regulator of erythroblast enucleation and terminal maturation suggesting FOXO3 modulation might be therapeutic in disorders with defective erythroid maturation.


Journal of Biological Chemistry | 2017

FOXO3 is Essential for Protecting Hematopoietic Stem and Progenitor Cells from Oxidative DNA Damage

Carolina L. Bigarella; Jianfeng Li; Pauline Rimmele; Raymond Liang; Robert W. Sobol; Saghi Ghaffari

Accumulation of damaged DNA in hematopoietic stem cells (HSC) is associated with chromosomal abnormalities, genomic instability, and HSC aging and might promote hematological malignancies with age. Despite this, the regulatory pathways implicated in the HSC DNA damage response have not been fully elucidated. One of the sources of DNA damage is reactive oxygen species (ROS) generated by both exogenous and endogenous insults. Balancing ROS levels in HSC requires FOXO3, which is an essential transcription factor for HSC maintenance implicated in HSC aging. Elevated ROS levels result in defective Foxo3−/− HSC cycling, among many other deficiencies. Here, we show that loss of FOXO3 leads to the accumulation of DNA damage in primitive hematopoietic stem and progenitor cells (HSPC), associated specifically with reduced expression of genes implicated in the repair of oxidative DNA damage. We provide further evidence that Foxo3−/− HSPC are defective in DNA damage repair. Specifically, we show that the base excision repair pathway, the main pathway utilized for the repair of oxidative DNA damage, is compromised in Foxo3−/− primitive hematopoietic cells. Treating mice in vivo with N-acetylcysteine reduces ROS levels, rescues HSC cycling defects, and partially mitigates HSPC DNA damage. These results indicate that DNA damage accrued as a result of elevated ROS in Foxo3−/− mutant HSPC is at least partially reversible. Collectively, our findings suggest that FOXO3 serves as a protector of HSC genomic stability and health.


British Journal of Haematology | 2016

Advances in understanding the mechanisms of erythropoiesis in homeostasis and disease

Raymond Liang; Saghi Ghaffari

Anaemia or decreased blood haemoglobin is the most common blood disorder often characterized by reduced red blood cell (RBC) numbers. RBCs are produced from differentiation and commitment of haematopoietic stem cells to the erythroid lineage by a process called erythropoiesis. Coordination of erythropoietin receptor signalling with several erythroid transcription factors including GATA1 is essential for this process. A number of additional players that are critical for RBC production have been identified in recent years. Major technological advances, such as the development of RNA interference, genetically modified animals, including zebrafish, and imaging flow cytometry have led to these discoveries; the emergence of ‐omics approaches in combination with the optimization of ex vivo erythroid cultures have also produced a more comprehensive understanding of erythropoiesis. Here we summarize studies describing novel regulators of erythropoiesis that modulate erythroid cell production in the context of human erythroid disorders involving hypoxia, iron regulation, immune‐related molecules, and the transcription factor FOXO3.


Journal of Bioenergetics and Biomembranes | 2017

Mitochondria and FOXO3 in stem cell homeostasis, a window into hematopoietic stem cell fate determination

Raymond Liang; Saghi Ghaffari

The production of all blood cells from hematopoietic stem cells (HSC) is highly sensitive to reactive oxygen species (ROS). Cumulating evidence suggests that mitochondria are critical for HSC fate determination. FOXO are known regulators of anti-oxidant response and key to the maintenance of HSC. Recent works indicate that FOXO3 is implicated in the control of mitochondrial function beyond regulating levels of ROS in HSC. Here we review these findings and discuss implications for homeostatic blood formation and stem cell fate determination.

Collaboration


Dive into the Raymond Liang's collaboration.

Top Co-Authors

Avatar

Saghi Ghaffari

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Carolina L. Bigarella

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Pauline Rimmele

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Dmitri Papatsenko

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Genís Campreciós

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Brigitte Izac

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Carlo Brugnara

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Erica K. Benson

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Jaehee Shim

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Luca Lambertini

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge