scholarly journals Hypoxia-Inducible Factor 1α Induces Fibrosis and Insulin Resistance in White Adipose Tissue

2009 ◽  
Vol 29 (16) ◽  
pp. 4467-4483 ◽  
Author(s):  
Nils Halberg ◽  
Tayeba Khan ◽  
Maria E. Trujillo ◽  
Ingrid Wernstedt-Asterholm ◽  
Alan D. Attie ◽  
...  

ABSTRACT Adipose tissue can undergo rapid expansion during times of excess caloric intake. Like a rapidly expanding tumor mass, obese adipose tissue becomes hypoxic due to the inability of the vasculature to keep pace with tissue growth. Consequently, during the early stages of obesity, hypoxic conditions cause an increase in the level of hypoxia-inducible factor 1α (HIF1α) expression. Using a transgenic model of overexpression of a constitutively active form of HIF1α, we determined that HIF1α fails to induce the expected proangiogenic response. In contrast, we observed that HIF1α initiates adipose tissue fibrosis, with an associated increase in local inflammation. “Trichrome- and picrosirius red-positive streaks,” enriched in fibrillar collagens, are a hallmark of adipose tissue suffering from the early stages of hypoxia-induced fibrosis. Lysyl oxidase (LOX) is a transcriptional target of HIF1α and acts by cross-linking collagen I and III to form the fibrillar collagen fibers. Inhibition of LOX activity by β-aminoproprionitrile treatment results in a significant improvement in several metabolic parameters and further reduces local adipose tissue inflammation. Collectively, our observations are consistent with a model in which adipose tissue hypoxia serves as an early upstream initiator for adipose tissue dysfunction by inducing a local state of fibrosis.

Blood ◽  
2011 ◽  
Vol 117 (2) ◽  
pp. 459-469 ◽  
Author(s):  
Chih-Chien Tsai ◽  
Yann-Jang Chen ◽  
Tu-Lai Yew ◽  
Ling-Lan Chen ◽  
Jir-You Wang ◽  
...  

Abstract Although low-density culture provides an efficient method for rapid expansion of human mesenchymal stem cells (MSCs), MSCs enriched by this method undergo senescence and lose their stem cell properties, which could be preserved by combining low-density and hypoxic culture. The mechanism was mediated through direct down-regulation of E2A-p21 by the hypoxia-inducible factor–1α (HIF-1α)–TWIST axis. Expansion under normoxia induced E2A and p21 expression, which were abrogated by overexpression of TWIST, whereas siRNA against TWIST up-regulated E2A and p21 in hypoxic cells. Furthermore, siRNA against p21 in normoxic cells enhanced proliferation and increased differentiation potential, whereas overexpression of p21 in hypoxic cells induced a decrease in proliferation and a loss of differentiation capacity. More importantly, MSCs expanded under hypoxic conditions by up to 100 population doublings, exhibited telomerase activity with maintained telomere length, normal karyotyping, and intact genetic integrity, and did not form tumors. These results support low-density hypoxic culture as a method for efficiently expanding MSCs without losing stem cell properties or increasing tumorigenicity.


2022 ◽  
Vol 12 (1) ◽  
Author(s):  
Shion Orikasa ◽  
Nobuyuki Kawashima ◽  
Kento Tazawa ◽  
Kentaro Hashimoto ◽  
Keisuke Sunada-Nara ◽  
...  

AbstractAccelerated dental pulp mineralization is a common complication in avulsed/luxated teeth, although the mechanisms underlying this remain unclear. We hypothesized that hypoxia due to vascular severance may induce osteo/odontoblast differentiation of dental pulp stem cells (DPSCs). This study examined the role of B-cell CLL/lymphoma 9 (BCL9), which is downstream of hypoxia-inducible factor 1α (HIF1α) and a Wnt/β-catenin transcriptional cofactor, in the osteo/odontoblastic differentiation of human DPSCs (hDPSCs) under hypoxic conditions. hDPSCs were isolated from extracted healthy wisdom teeth. Hypoxic conditions and HIF1α overexpression induced significant upregulation of mRNAs for osteo/odontoblast markers (RUNX2, ALP, OC), BCL9, and Wnt/β-catenin signaling target genes (AXIN2, TCF1) in hDPSCs. Overexpression and suppression of BCL9 in hDPSCs up- and downregulated, respectively, the mRNAs for AXIN2, TCF1, and the osteo/odontoblast markers. Hypoxic-cultured mouse pulp tissue explants showed the promotion of HIF1α, BCL9, and β-catenin expression and BCL9-β-catenin co-localization. In addition, BCL9 formed a complex with β-catenin in hDPSCs in vitro. This study demonstrated that hypoxia/HIF1α-induced osteo/odontoblast differentiation of hDPSCs was partially dependent on Wnt/β-catenin signaling, where BCL9 acted as a key mediator between HIF1α and Wnt/β-catenin signaling. These findings may reveal part of the mechanisms of dental pulp mineralization after traumatic dental injury.


2006 ◽  
Vol 70 (6) ◽  
pp. 1856-1865 ◽  
Author(s):  
Ji-Hong Lim ◽  
Jong-Wan Park ◽  
Myung-Suk Kim ◽  
Sang-Ki Park ◽  
Randall S. Johnson ◽  
...  

2009 ◽  
Vol 424 (1) ◽  
pp. 143-151 ◽  
Author(s):  
Tetsuhiro Tanaka ◽  
Michael Wiesener ◽  
Wanja Bernhardt ◽  
Kai-Uwe Eckardt ◽  
Christina Warnecke

HIF (hypoxia-inducible factor)-3α is the third member of the HIF transcription factor family. Whereas HIF-1α and -2α play critical roles in the cellular and systemic adaptation to hypoxia, little is known about the regulation and function of HIF-3α. At least five different splice variants may be expressed from the human HIF-3α locus that are suggested to exert primarily negative regulatory effects on hypoxic gene induction. In the present paper, we report that hypoxia induces the human HIF-3α gene at the transcriptional level in a HIF-1-dependent manner. HIF-3α2 and HIF-3α4 transcripts, the HIF-3α splice variants expressed in Caki-1 renal carcinoma cells, rapidly increased after exposure to hypoxia or chemical hypoxia mimetics. siRNA (small interfering RNA)-mediated HIF-α knockdown demonstrated that HIF-3α is a specific target gene of HIF-1α, but is not affected by HIF-2α knockdown. In contrast with HIF-1α and HIF-2α, HIF-3α is not regulated at the level of protein stability. HIF-3α protein could be detected under normoxia in the cytoplasm and nuclei, but increased under hypoxic conditions. Promoter analyses and chromatin immunoprecipitation experiments localized a functional hypoxia-responsive element 5′ to the transcriptional start of HIF-3α2. siRNA-mediated knockdown of HIF-3α increased transactivation of a HIF-driven reporter construct and mRNA expression of lysyl oxidase. Immunohistochemistry revealed an overlap of HIF-1α-positive and HIF-3α-positive areas in human renal cell carcinomas. These findings shed light on a novel aspect of HIF-3α as a HIF-1 target gene and point to a possible role as a modulator of hypoxic gene induction.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2586-2586
Author(s):  
Rodrigo Jacamo ◽  
Juliana Benito ◽  
Olga Frolova ◽  
Ye Chen ◽  
Hongbo Lu ◽  
...  

Abstract Abstract 2586 Resistance to chemotherapy can be mediated by genetic, epigenetic and microenvironmental causes. Only recently the connection between leukemia growth and survival and the hypoxic state of the BM microenvironment has been appreciated, by work conducted by us and others (Fiegl M et.al. Blood 2009; 113: 1504–1512; Harrison JS et. al., Blood 2002; 99). In extension of this concept we investigated the role of Hypoxia-Inducible-Factor 1α (HIF1A), the master regulator of hypoxia induced responses, in the microenvironment and its relevance for leukemia progression. Here we focused on the role of hypoxia and HIF transcription factors in cells contributing to the BM microenvironment, the mesenchymal stromal cells (MSC). Co-culture of lymphoid (NALM6) and myeloid (OCI-AML3) leukemic cell lines with BM-derived MSC under hypoxic conditions (1% O2) stimulated the secretion of a number of pro-survival cytokines and chemokines (including IL-6, VEGF, Beta-NGF and SDF-1α) that were quantified in co-culture supernatants by Luminex flow cytometry (Table 1). These findings suggest that hypoxia, and possibly its main mediator, the transcription factor HIF1A, may be responsible for the increased production of these factors. Since the chemokine stromal cell-derived factor-1α (SDF-1α) is involved in the attraction of leukemic cells towards cells of the BM microenvironment, we next investigated the role of HIF1A expression in MSC and its effect on SDF-1 secretion and migration of leukemic cells under hypoxic conditions. To this end, we generated primary human BM MSC stably transduced with lentiviral-encoded shRNA against HIF1A. SDF-1α transcription levels measured by qRT-PCR were diminished (∼30%, p<0.01) in HIF1A-silenced MSCs compared to control MSCs expressing non-silencing shRNA. This correlated with significantly reduced transwell migration of OCI-AML3 cells towards HIF1A-silenced MSCs compared with control (non-silencing) MSCs (∼35%, p<0.05) under hypoxic conditions. We next examined the contribution of hypoxia and HIF1A in the protective role of the BM microenvironment against standard chemotherapy with AraC and Doxorubicin. To this end, we performed in vitro experiments co culturing OCI-AML3 cells with either HIF1A-silenced MSCs or control MSCs under hypoxic conditions. After 48h of drug treatment a significant decrease in chemotherapy-induced apoptosis in leukemic cells co-cultured with control MSCs compared to leukemic cells cultured alone was observed. In turn, chemoresistance was reduced in OCI-AML3 co-cultured with HIF1A-silenced MSC, suggesting that hypoxia mediates chemoresistance largely through its effects on cells of the BM microenvironment. It has been shown that leukemic cells seem to exhibit increased dependency on glycolysis for ATP generation, which is frequently associated with resistance to therapeutic agents. Therefore, we measured the production of lactic acid (LA) in leukemic cells co-cultured with MSC in hypoxia compared to normoxia. In agreement with previous observations, we found that REH and primary ALL cells produced more LA when they were co-cultured with MSC under hypoxia compared to normoxia (∼1.8 fold, p<0.05). When REH cells were co-cultured with HIF1A-silenced MSCs in hypoxic conditions the lactic acid production was slightly but significantly reduced (∼20%, p<0.05) compared with the values observed in REH-control MSCs co-culture supernatants. Altogether, these findings strongly point to hypoxia and HIF1A as pivotal components in the protection from chemotherapy mediated by the BM microenvironment. We propose that targeting HIF1A and hypoxia in the protective cells of the bone marrow niches may represent a new approach to increase chemosensitivity of leukemic cells and hopefully improve the existing therapeutic strategies. Table 1: Fold increase observed in leukemic cells-MSC co-culture supernatants in hypoxia compared to normoxia. OCI-AML3+MSC NALM6+MSC IL-6 ∼3.1 ∼1.2 VEGF ∼3 ∼2 B-NGF ∼8 ∼10 SDF-1 ∼1.5 ∼1.5 Disclosure: No relevant conflicts of interest to declare.


Epigenetics ◽  
2014 ◽  
Vol 9 (4) ◽  
pp. 600-610 ◽  
Author(s):  
Laura Tudisco ◽  
Floriana Della Ragione ◽  
Valeria Tarallo ◽  
Ivana Apicella ◽  
Maurizio D'Esposito ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document