Proton export alkalinizes intracellular pH and reprograms carbon metabolism to drive hematopoietic progenitor growth

Blood ◽  
2021 ◽  
Author(s):  
Cheuk Him Man ◽  
Francois Emile Mercier ◽  
Nian Liu ◽  
Wentao Dong ◽  
Gregory Stephanopoulos ◽  
...  

Proton export is often considered a detoxifying process in animal cells with monocarboxylate symporters co-exporting excessive lactate and protons during glycolysis or the Warburg effect. Here we report a novel mechanism by which lactate/H+ export is sufficient to induce cell growth. Increased lactate/proton export induces intracellular alkalization that selectively activates catalysis by key metabolic gatekeeper enzymes, HK1/PKM2/G6PDH, thereby enhancing glycolytic and pentose phosphate pathway carbon flux. The result is increased nucleotide levels, NADPH/NADP+ ratio and cell proliferation. Simply increasing the lactate/proton symporter, MCT4, or sodium-proton antiporter, NHE1 was sufficient to increase intracellular-pH (pHi) and give normal hematopoietic cells a significant competitive growth advantage in vivo. This process does not require additional cytokine triggers and is exploited in malignancy where leukemogenic mutations epigenetically increase MCT4. Inhibiting MCT4 decreased intracellular pH, carbon flux and eliminated acute myeloid leukemia-initiating-cells without cytotoxic chemotherapy. Intracellular alkalization is a primitive mechanism by which proton partitioning can directly reprogram carbon metabolism for cell growth.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3765-3765
Author(s):  
Cheuk-Him Man ◽  
David T. Scadden ◽  
Francois Mercier ◽  
Nian Liu ◽  
Wentao Dong ◽  
...  

Acute myeloid leukemia (AML) cells exhibit metabolic alterations that may provide therapeutic targets not necessarily evident in the cancer cell genome. Among the metabolic features we noted in AML compared with normal hematopoietic stem and progenitors (HSPC) was a strikingly consistent alkaline intracellular pH (pHi). Among candidate proton regulators, monocarboxylate transporter 4 (MCT4) mRNA and protein were differentially increased in multiple human and mouse AML cell lines and primary AML cells. MCT4 is a plasma membrane H+and lactate co-transporter whose activity necessarily shifts protons extracellularly as intracellular lactate is extruded. MCT4 activity is increased when overexpressed or with increased intracellular lactate generated by glycolysis in the setting of nutrient abundance. With increased MCT4 activity, extracellular lactate and protons will increase causing extracellular acidification while alkalinizing the intracellular compartment. MCT4-knockout (MCT4-KO) of mouse and human AMLdid not induce compensatory MCT1 expression, reduced pHi, suppressed proliferation and improved animal survival. Growth reduction was experimentally defined to be due to intracellular acidification rather than lactate accumulation by independent modulation of those parameters. MCT4-KOmetabolic profiling demonstrated decreased ATP/ADP and increased NADP+/NADPH suggesting suppression of glycolysis and the pentose phosphate pathway (PPP) that was confirmed by stable isotopic carbon flux analyses. Notably,the enzymatic activity of purified gatekeeper enzymes, hexokinase 1 (HK1), pyruvate kinase M2 isoform (PKM2) and glucose-6-phosphate dehydrogenase (G6PDH) was sensitive to pH with increased activity at the leukemic pHi (pH 7.6) compared to normal pHi (pH 7.3). Evaluating MCT4 transcriptional regulation, we defined that activating histonemarks, H3K27ac and H3K4me3, were enriched at the MCT4 promoter region as were transcriptional regulators MLL1 and Brd4 by ChIP in AML compared with normal cells. Pharmacologic inhibition of Brd4 suppressed Brd4 and H3K27ac enrichment and MCT4 expression in AML and reduced leukemic cell growth. To determine whether MCT4 based pHi changes were sufficient to increase cell proliferation, we overexpressed MCT4 in normal HSPC and demonstrated in vivo increases in growth in conjunction with pHi alkalization. Some other cell types also were increased in their growth kinetics by MCT4 overexpression and pHi increase. Therefore, proton shifting may be a means by which cells respond to nutrient abundance, co-transporting lactate and protons out of the cell, increasing the activity of enzymes that enhance PPP and glycolysis for biomass generation. Epigenetic changes in AML appear to exploit that process by increasing MCT4 expression to enforce proton exclusion thereby gaining a growth advantage without dependence on signaling pathways. Inhibiting MCT4 and intracellular alkalization may diminish the ability of AML to outcompete normal hematopoiesis. Figure Disclosures Scadden: Clear Creek Bio: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Novartis: Other: Sponsored research; Editas Medicine: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Bone Therapeutics: Consultancy; Fog Pharma: Consultancy; Red Oak Medicines: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; LifeVaultBio: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Magenta Therapeutics: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Agios Pharmaceuticals: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Fate Therapeutics: Consultancy, Equity Ownership.


2015 ◽  
Vol 26 (7) ◽  
pp. 763-773
Author(s):  
Muhammad N. Aslam ◽  
Shannon McClintock ◽  
Shazli P. Khan ◽  
Patricia Perone ◽  
Ronald Allen ◽  
...  

1989 ◽  
Vol 67 (12) ◽  
pp. 2994-3004 ◽  
Author(s):  
Patrick J. Walsh ◽  
C. Louise Milligan

This review discusses the mechanisms by which animal cells regulate intracellular pH (pHi), the variations in pHi encountered in vivo, and the impact that variations in pHi (and other acid–base variables) have on metabolism. Cells regulate pHi by a combination of (i) physicochemical buffering by intracellular components; (ii) transport of acids and bases across the plasma membrane; and (iii) production and consumption of acids and bases by metabolism. Ionic transport is by far the best studied of these three mechanisms, and several specific plasma membrane exchangers (e.g., Na+–H+ exchange) are important regulators of pHi The precise quantitative contribution of the other two mechanisms to pHi regulation awaits further study. Intracellular pH variations in vivo can be substantial (i.e., up to 1 unit in some cases) and can lead to marked changes in metabolism. Furthermore, changes in carbon dioxide tension and bicarbonate concentration can also affect metabolism. Catecholamines appear to be important regulatory signals in metabolic compensation for acid–base perturbations, but in some cases acid–base disturbances may produce adaptive metabolic changes directly.


Metabolites ◽  
2020 ◽  
Vol 10 (7) ◽  
pp. 285 ◽  
Author(s):  
Khalid O. Alfarouk ◽  
Samrein B. M. Ahmed ◽  
Robert L. Elliott ◽  
Amanda Benoit ◽  
Saad S. Alqahtani ◽  
...  

The Pentose Phosphate Pathway (PPP) is one of the key metabolic pathways occurring in living cells to produce energy and maintain cellular homeostasis. Cancer cells have higher cytoplasmic utilization of glucose (glycolysis), even in the presence of oxygen; this is known as the “Warburg Effect”. However, cytoplasmic glucose utilization can also occur in cancer through the PPP. This pathway contributes to cancer cells by operating in many different ways: (i) as a defense mechanism via the reduced form of nicotinamide adenine dinucleotide phosphate (NADPH) to prevent apoptosis, (ii) as a provision for the maintenance of energy by intermediate glycolysis, (iii) by increasing genomic material to the cellular pool of nucleic acid bases, (iv) by promoting survival through increasing glycolysis, and so increasing acid production, and (v) by inducing cellular proliferation by the synthesis of nucleic acid, fatty acid, and amino acid. Each step of the PPP can be upregulated in some types of cancer but not in others. An interesting aspect of this metabolic pathway is the shared regulation of the glycolytic and PPP pathways by intracellular pH (pHi). Indeed, as with glycolysis, the optimum activity of the enzymes driving the PPP occurs at an alkaline pHi, which is compatible with the cytoplasmic pH of cancer cells. Here, we outline each step of the PPP and discuss its possible correlation with cancer.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1173-1173
Author(s):  
Shanshan Suo ◽  
Le Xuan Truong Nguyen ◽  
Fenglin Li ◽  
Dandan Zhao ◽  
Junjing Qiao ◽  
...  

Abstract Acute myeloid leukemia (AML) is a devastating hematopoietic malignancy. With current therapies, only approximately 30% of patients achieve long-term survival. Therefore, novel, more active and less toxic treatments are urgently needed. Programmed death-1 (PD-1) is a cell surface receptor that functions as a T cell checkpoint and plays a central role in regulating T cell exhaustion. Binding of PD-1 to its ligand, programmed death-ligand 1 (PD-L1), activates downstream signaling pathways and inhibits T cell activation. Abnormally high PD-L1 expression on tumor cells and antigen-presenting cells in the tumor microenvironment mediates tumor immune escape, and PD-1/PD-L1 immune checkpoint blockade has showed promising results in cancer patients. Recently, PD-1 expressed on melanoma cells was also shown to play a pivotal role in tumor growth. To date, in AML, the function of PD-1 has been mainly studied in the host T cells, while little is known regarding the role of PD-1 in AML cells. Herein, we examined the level and role of PD-1 in AML cells using AML murine model and patient samples. We used MLL PTD/WT/Flt3 ITD/ITD knock-in mouse in B6 background, a well characterized AML model, to study the expression and function of PD-1 in AML. Flow cytometric analysis of LSK (Lin -Sca-l1 +c-kit +) cells from the bone marrow (BM) of wild-type (WT, n=5) and AML (n=10) mice showed that 20.9%-61.9% of AML LSKs versus (vs) <5.0% of normal LSKs are PD-1 positive (P< 0.0001). Western blot and Q-RT-PCR analysis confirmed higher levels of PD-1 in AML LSKs than in normal LSKs. The PD-1 levels on LSKs increased over time and associated with disease progression. Similar results were obtained in AML patients, showing PD-1 + in 0.2%-14.6% of AML CD34 + cells vs < 1.0% of normal CD34 + cells (P< 0.05). Data analysis based on TCGA showed that higher PD-1 levels are associated with shorter survival in AML patients (P=0.0125). To assess the functional role of PD-1 in AML, we sorted PD-1 + and PD-1 - fractions fromAML LSKs and observed a lower frequency of quiescent cells (G0, 16.60% vs 44.87%, P< 0.05) and a higher cell growth rate in the PD-1 + vs PD-1 - cells. Further in vivo study showed that PD-1 + AML LSKs (CD45.2) generated higher white blood cell (WBC) counts (P< 0.0001), higher AML engraftment (P< 0.0001) and a shorter survival (median survival 57.5 vs >75 days, P< 0.001) in recipient mice (CD45.1) compared with PD-1 - AML LSKs. Similar results were observed in human samples. Compared to PD-1 - CD34 + cells, PD-1 + CD34 + cells are less quiescent and more proliferative (P< 0.01). PD-1 + AML blasts had higher engraftment rate (13.18% vs 2.68%, p=0.0002) and shorter survival (median survival: 27 vs 45 days, P= 0.0008) in NSGS mice than PD-1 - AML blasts. To evaluate if these in vivo differences observed in PD-1 + vs PD-1 - AML LSKs were mediated by interactions between PD-1 + AML and T cells, PD-1 + and PD-1 -LSKs from AML mouse were transplanted into T-, B- cell-deficient NSG mice. Recipient mice transplanted with PD-1 + AML LSKs had higher WBC counts (P< 0.01), higher AML engraftment (P< 0.0001) and a shorter survival (median survival: 76 vs >130 days, P< 0.0001) than recipients with PD-1 - AML LSKs, suggesting that these differences were T cell-independent. Next, we examined whether blocking PD-1 could affect leukemic cell growth. We sorted LSK cells from AML mice and performed colony forming cell (CFC) assay in the presence of anti-PD1 antibody or isotype antibody. Blocking PD-1 with anti-PD-1 antibody significantly suppressed CFC and cell growth in vitro but did not induce apoptosis compared to isotype control antibody. To explore the molecular mechanism by which PD-1 contributes to AML growth, we then sorted PD-1 + and PD-1 - LSKs from AML mice for molecular studies. Western blot assays revealed higher levels of SHP-2 and phosphorylated (p) -ERK in PD-1 + vs PD-1 - AML LSKs. We validated these results in primary human AML cells by immunofluorescence staining. Confocal microscopy of PD-1 + and PD-1 - human AML CD34 +cells demonstrated that PD-1 localized at the cell membrane and in the cytoplasm and p-ERK was markedly enhanced in the PD-1 + CD34 + cells. In conclusion, we showed here that a subpopulation of murine and human AML blasts expresses high levels of PD-1 which mediated disease initiation and growth through activation of the MAPK/ERK signaling pathways. PD-1 blocking antibody reversed these activities and might contribute to the clinical efficacy of anti-PD-1 therapy in AML. Disclosures Marcucci: Novartis: Other: Speaker and advisory scientific board meetings; Agios: Other: Speaker and advisory scientific board meetings; Abbvie: Other: Speaker and advisory scientific board meetings.


2021 ◽  
Author(s):  
Thomas Wieloch ◽  
Angela Augusti ◽  
Juergen Schleucher

As the central carbon uptake pathway in photosynthetic cells, the Calvin-Benson cycle is among the most important biochemical cycles for life on Earth. Recently, anaplerotic carbon flux (through the chloroplast-localised oxidative branch of the pentose phosphate pathway) into this cycle was proposed. Here, we measured intramolecular deuterium abundances in leaf starch of Helianthus annuus grown at varying ambient CO2 concentrations, Ca. Additionally, we modelled deuterium fractionations expected for the anaplerotic pathway and compared modelled with measured fractionations. We report deuterium fractionation signals at starch glucose H1 and H2. Below a response change point, these signals increase with decreasing Ca consistent with modelled fractionations by anaplerotic flux. Under normal growth conditions (Ca≥450 ppm corresponding to intercellular CO2 concentrations, Ci, ≥328 ppm), we estimate negligible anaplerotic flux. At Ca=180 ppm (Ci=140 ppm), we estimate that of the glucose 6-phosphate entering the starch biosynthesis pathway more than 11.5% is diverted into the anaplerotic pathway. In conclusion, we report evidence consistent with anaplerotic carbon flux into the Calvin-Benson cycle in vivo. We propose the flux may help to (i) maintain high levels of ribulose 1,5-bisphosphate under source-limited growth conditions to facilitate photorespiratory nitrogen assimilation required to build-up source strength and (ii) counteract oxidative stress.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2049-2049 ◽  
Author(s):  
D Ball Edward ◽  
Bruno C. Medeiros ◽  
Larissa Balaian ◽  
Tracy Roque ◽  
Sue Corringham ◽  
...  

Abstract Abstract 2049 Poster Board II-26 Acute myeloid leukemia (AML) cells express the cell surface antigen CD33 that, upon ligation with a monoclonal antibody (mAb), is a downregulator of cell growth in a Syk-dependent manner. An anti-CD33 mAb coupled to calechiamycin, gemtuzumab ozogamicin (GO), is used for the treatment of AML. We demonstrated that the response of AML cells to GO treatment also depends on Syk and SHP-1 expression (Leukemia 20:2093, 2006). Syk upregulation by the demethylating agent 5-azacytidine (5-aza) induced re-expression of Syk in some cases, resulting in enhanced GO and anti-CD33-mediated inhibition of leukemia cell growth. Thus, the cytotoxicity of both GO and anti-CD33 in primary AML samples was associated with Syk expression. 5-Aza restored Syk and increased the sensitivity of originally Syk-negative, non-responsive cells to CD33 ligation to levels of Syk-positive cells. We designed a clinical trial examining if treatment with 5-aza prior to GO is safe, efficacious, and whether in vivo responses to GO correlated with Syk expression and induction by 5-aza. Here we report the interim results of this trial. In Phase I, 14 patients (9 males [7 Caucasian, 1 Asian, 1 Hispanic], 5 females [2 Caucasian, 3 Asian], age range: 39-82 years [median: 66]) were treated with 75mg/m2 5-aza and GO in a dose-escalation manner: the first cohort (n=3) received 5-aza for 2 days followed by GO at 3 mg/m2 on days 3 and 17; the second cohort (n=3) received 5-aza for 2 days followed by GO at 6 mg/m2 on days 3 and 17; the third cohort (n=4) received 5-aza for 4 days followed by GO at 6 mg/m2 on days 5 and 19; and the fourth cohort (n=4) at 5-aza for 6 days followed by GO at 6 mg/m2 on days 7 and 21. No dose-limiting toxicities have been encountered. There were no responses in the first two cohorts (5 patients in 1st rel, 1 in 2nd rel at start of treatment); one patient in cohort 3 achieved a CR (in 1st relapse), two in cohort 4 have achieved CR (1 in 1st rel and 1 in 2nd rel). We are in Phase II of the study: Cohort 4 dose 5-aza for 6 days followed by GO at 6 mg/m2 on days 7 and 21. Patients remained on study until ANC recovered to 1000/mm3 for 3 consecutive days and were assessed for response to treatment. Average time to ANC recovery (n=6) 30 days (range 15-42) (median 33 days). Average length of time on study: 46 days. Prior to therapy, Syk was expressed in 10 of the 13 cases (baseline data not available on one case). After 5-aza treatment, Syk was re-expressed in each of the 3 negative cases, and increased over baseline in one case that was previously Syk +. SHP-1 was positive in 10 of the 13 cases and was re-expressed in each of the 3 negative cases. The leukemia cells from the patients who achieved CR were Syk+ in 2 of 3 cases. Syk was re-expressed in the one negative case after 5-aza. SHP-1 was expressed in 2 of 3 cases at baseline, and re-expressed in the one negative case after 5-aza. In vitro we analyzed inhibition of proliferation (for patients 1-6) or colony formation (for patients 7-14) induced by 5-aza and GO treatment. In all cases 5-azacytidine alone mediated about 50%, and GO alone about 40% cytotoxicity. Together they killed about 80% of leukemia cells. We also compared pre- and post 5-aza samples from the same patients: in all cases 5-aza treatment increased the GO-mediated cytotoxicity. These initial data show that in vivo exposure to 5-aza can induce the expression of two biomarkers involved in the intracellular response to GO. Correlations with response will be made in the Phase II portion of the study now underway. Disclosures: Ball: Celgene: Research Funding. Off Label Use: 5-azacytidine used in treatment of AML.


2020 ◽  
Vol 13 (1) ◽  
pp. 293-314 ◽  
Author(s):  
Zhuo Zhang ◽  
Xiawei Cheng ◽  
Yuzheng Zhao ◽  
Yi Yang

As the core component of cell metabolism, central carbon metabolism, consisting of glycolysis, the pentose phosphate pathway, and the tricarboxylic acid cycle converts nutrients into metabolic precursors for biomass and energy to sustain the life of virtually all extant species. The metabolite levels or distributions in central carbon metabolism often change dynamically with cell fates, development, and disease progression. However, traditional biochemical methods require cell lysis, making it challenging to obtain spatiotemporal information about metabolites in living cells and in vivo. Genetically encoded fluorescent sensors allow the rapid, sensitive, specific, and real-time readout of metabolite dynamics in living organisms, thereby offering the potential to fill the gap in current techniques. In this review, we introduce recent progress made in the development of genetically encoded fluorescent sensors for central carbon metabolism and discuss their advantages, disadvantages, and applications. Moreover, several future directions of metabolite sensors are also proposed.


2006 ◽  
Vol 175 (4S) ◽  
pp. 257-257
Author(s):  
Jennifer Sung ◽  
Qinghua Xia ◽  
Wasim Chowdhury ◽  
Shabana Shabbeer ◽  
Michael Carducci ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document